1
|
Mane V, Mehta R, Alvarez N, Sharma V, Park S, Fox A, DeCarlo C, Yang X, Perlin DS, Powell RLR. In vivo antiviral efficacy of LCTG-002, a pooled, purified human milk secretory IgA product, against SARS-CoV-2 in a murine model of COVID-19. Hum Vaccin Immunother 2024; 20:2303226. [PMID: 38251677 PMCID: PMC10807469 DOI: 10.1080/21645515.2024.2303226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 01/05/2024] [Indexed: 01/23/2024] Open
Abstract
Immunoglobulin A (IgA) is the most abundant antibody (Ab) in human mucosae, with secretory form (sIgA) being dominant and uniquely stable. sIgA is challenging to produce recombinantly but is naturally found in human milk, which could be considered a global resource for this biologic, justifying its development as a mucosal therapeutic. Presently, SARS-CoV-2 was utilized as a model mucosal pathogen, and methods were developed to efficiently extract human milk sIgA from donors who were naïve to SARS-CoV-2 or had recovered from infection that elicited high-titer anti-SARS-CoV-2 Spike sIgA in their milk (pooled to make LCTG-002). Mass spectrometry determined that proteins with a relative abundance of 1% or greater were all associated with sIgA. Western blot demonstrated that all batches consisted predominantly of sIgA. Compared to control IgA, LCTG-002 demonstrated significantly higher Spike binding (mean endpoint of 0.87 versus 5.87). LCTG-002 was capable of blocking the Spike receptor-binding domain - angiotensin-converting enzyme 2 (ACE2) interaction with significantly greater potency compared to control (mean LCTG-002 IC50 154ug/mL versus 50% inhibition not achieved for control), and exhibited significant neutralization activity against Spike-pseudotyped virus infection (mean LCTG-002 IC50 49.8ug/mL versus 114.5ug/mL for control). LCTG-002 was tested for its capacity to reduce viral lung burden in K18+hACE2 transgenic mice inoculated with SARS-CoV-2. LCTG-002 significantly reduced SARS-CoV-2 titers compared to control when administered at 0.25 mg/day or 1 mg/day, with a maximum TCID50 reduction of 4.9 logs. This innovative study demonstrates that LCTG-002 is highly pure and efficacious in vivo, supporting further development of milk-derived, polyclonal sIgA therapeutics.
Collapse
Affiliation(s)
- Viraj Mane
- Lactiga US, Inc. 675 US-1, North Brunswick, NJ, USA
| | - Rikin Mehta
- Lactiga US, Inc. 675 US-1, North Brunswick, NJ, USA
| | - Nadine Alvarez
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
| | - Vijeta Sharma
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
| | - Steven Park
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
| | - Alisa Fox
- Icahn School of Medicine at Mount Sinai, Department of Medicine, Division of Infectious Diseases, New York, NY, USA
| | - Claire DeCarlo
- Icahn School of Medicine at Mount Sinai, Department of Medicine, Division of Infectious Diseases, New York, NY, USA
| | - Xiaoqi Yang
- Icahn School of Medicine at Mount Sinai, Department of Medicine, Division of Infectious Diseases, New York, NY, USA
| | - David S. Perlin
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
| | - Rebecca L. R. Powell
- Icahn School of Medicine at Mount Sinai, Department of Medicine, Division of Infectious Diseases, New York, NY, USA
| |
Collapse
|
2
|
Di Stefano M, Mirabella L, Cotoia A, Faleo G, Rauseo M, Rizzo AC, Fiore JR, Cinnella G, Serviddio G. A Possible Protective Effect of IgA Against Severe Acute Respiratory Syndrome Corona Virus 2 (SARS-CoV-2) in Bronchoalveolar Lavage in COVID-19 Patients Admitted to Intensive Care Unit. Viruses 2024; 16:1851. [PMID: 39772161 PMCID: PMC11680283 DOI: 10.3390/v16121851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025] Open
Abstract
SARS-CoV-2 infection induces a humoral immune response, producing virus-specific antibodies such as IgM, IgG, and IgA. IgA antibodies are present at mucosal sites, protecting against respiratory and other mucosal infections, including SARS-CoV-2, by neutralizing viruses or impeding attachment to epithelial cells. Since SARS-CoV-2 spreads through the nasopharynx, the specific IgAs of SARS-CoV-2 are produced quickly after infection, effectively contributing to virus neutralization. Dimeric IgA has been reported to be 10 to 15 times more potent than its equivalent IgG, suggesting that this isotype may be particularly interesting in developing new monoclonal antibodies and/or new vaccines efficiently neutralizing the virus at the mucosal sites. It is still unclear whether IgA antibodies in BAL might play a role in the disease course and if their presence may have a prognostic significance. However, a harmful effect on diseases with high IgA titers has been reported. This study evaluated mucosal-specific IgA and IgG profiles in BAL of patients with COVID-19 acute respiratory failure admitted to the ICU. We included 57 patients (41 males and 16 females), admitted to the ICU of the University of Foggia. We used a commercially available ELISA assay to evaluate the presence of SARS-CoV-2 IgG and IgA antibodies in plasma and BAL of the 57 hospitalized patients with severe COVID-19 respiratory failure. However, 40/57 BAL and plasma from infected patients were available for the ELISA test; the remaining specimens were unsuitable. IgG and IgA antibodies against SARS-CoV-2 were detectable in 37 (92.5%) and 40 (100%) plasma specimens, respectively. IgG antibodies were found in a single sample, while IgAs were detected in 19 of 40 BAL samples analyzed. Correlations between these parameters and patient outcomes reveal a signature associated with survival. Interestingly, a statistically significant inverse correlation was found between the mortality rate and the presence of IgA to SARS-CoV-2 in BAL specimens. None of the 19 patients with a positive IgA died, compared to 7 out of 12 patients with a negative IgA-BAL (p: <0.0004). Despite being limited in size, this study suggests a significant protective effect of mucosal immunity in COVID-19 patients, even in advanced disease stages, and a role of IgA in the defense against the virus, as well as the possible use of effective vaccines and therapeutic strategies based on IgA antibodies.
Collapse
Affiliation(s)
- Mariantonietta Di Stefano
- Department of Surgical and Medical Science, Section of Infectious Diseases, University of Foggia, 71122 Foggia, Italy; (G.F.); (J.R.F.)
| | - Lucia Mirabella
- Anesthesia and Intensive Care Unit, Department of Surgical and Medical Science, Policlinico Riuniti di Foggia, University of Foggia, 71122 Foggia, Italy; (L.M.); (A.C.); (M.R.); (A.C.R.); (G.C.)
| | - Antonella Cotoia
- Anesthesia and Intensive Care Unit, Department of Surgical and Medical Science, Policlinico Riuniti di Foggia, University of Foggia, 71122 Foggia, Italy; (L.M.); (A.C.); (M.R.); (A.C.R.); (G.C.)
| | - Giuseppina Faleo
- Department of Surgical and Medical Science, Section of Infectious Diseases, University of Foggia, 71122 Foggia, Italy; (G.F.); (J.R.F.)
| | - Michela Rauseo
- Anesthesia and Intensive Care Unit, Department of Surgical and Medical Science, Policlinico Riuniti di Foggia, University of Foggia, 71122 Foggia, Italy; (L.M.); (A.C.); (M.R.); (A.C.R.); (G.C.)
| | - Anna Chiara Rizzo
- Anesthesia and Intensive Care Unit, Department of Surgical and Medical Science, Policlinico Riuniti di Foggia, University of Foggia, 71122 Foggia, Italy; (L.M.); (A.C.); (M.R.); (A.C.R.); (G.C.)
| | - Josè Ramon Fiore
- Department of Surgical and Medical Science, Section of Infectious Diseases, University of Foggia, 71122 Foggia, Italy; (G.F.); (J.R.F.)
| | - Gilda Cinnella
- Anesthesia and Intensive Care Unit, Department of Surgical and Medical Science, Policlinico Riuniti di Foggia, University of Foggia, 71122 Foggia, Italy; (L.M.); (A.C.); (M.R.); (A.C.R.); (G.C.)
| | - Gaetano Serviddio
- C.U.R.E. (University Center for Liver Disease Research and Treatment), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy;
| |
Collapse
|
3
|
Wang X, Xiao C, Wu S, Lin Q, Lin S, Liu J, Ye D, Wang C, Guo P. Impacts of Nano-Composite of Copper and Carbon on Intestinal Luminal Micro-Ecosystem and Mucosal Homeostasis of Yellow-Feather Broilers. Microorganisms 2024; 12:2247. [PMID: 39597636 PMCID: PMC11596944 DOI: 10.3390/microorganisms12112247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
The present study was undertaken to evaluate the impacts of nano-composites of copper and carbon (NCCC) on the intestinal luminal micro-ecosystem and mucosal homeostasis of yellow-feather broilers. A total of two-hundred and forty 1-day-old male yellow-feather broilers were randomly allocated into four groups, each with five replications of twelve birds. The control (CON) group received a corn-soybean basal diet, while the N50, N100, and N200 groups were supplemented with 50, 100, and 200 mg/kg of NCCC in basal diets, respectively. The trial duration was 63 days. The findings demonstrated that there were slight impacts of NCCC addition on the intestinal luminal micro-ecosystem of broilers, with the fecal moisture content in the N100 group being slightly higher on Day 3 in the starter phase (p < 0.05). The cecal microbiota structure also did not obviously change (p > 0.05), in spite of the fall in the relative abundance of the Ruminococcus torques group in the N50 group and norank Clostridia UCG-014 in N200 group (p < 0.05). But for intestinal mucosal homeostasis, NCCC played a crucial part in jejunal morphology, tight junction, immunologic status, and antioxidant capacity. There was linear growth in villus height and a quadratic increase in villus height, crypt depth and their ratio with the increase in NCCC dosage (p < 0.05), and 100 mg/kg NCCC supplementation could intensify the expression of CLDN-3 genes (p < 0.05). In addition, IL-4 and IL-10 linearly increased after NCCC treatment (p < 0.05), along with some irregular changes in sIgA (p < 0.05). In addition, higher jejunal mucosal total antioxidant capacities in N50 and N200 groups were also observed (p < 0.05). Overall, NCCC treatment optimized the intestinal mucosa function of broilers in terms of physical barrier and immune and antioxidant capacities, but exerted subtle influence in the luminal environment of yellow-feather broilers. More precisely, dietary supplementation with 50 mg/kg NCCC is recommended for intestinal homeostasis of broilers.
Collapse
Affiliation(s)
- Xianglin Wang
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (X.W.); (C.X.); (S.W.); (Q.L.); (S.L.); (C.W.)
| | - Chunlong Xiao
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (X.W.); (C.X.); (S.W.); (Q.L.); (S.L.); (C.W.)
| | - Shuqing Wu
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (X.W.); (C.X.); (S.W.); (Q.L.); (S.L.); (C.W.)
| | - Qingjie Lin
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (X.W.); (C.X.); (S.W.); (Q.L.); (S.L.); (C.W.)
| | - Shiying Lin
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (X.W.); (C.X.); (S.W.); (Q.L.); (S.L.); (C.W.)
| | - Jing Liu
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou 350013, China; (J.L.); (D.Y.)
- Livestock and Poultry Genetic Breeding Key Laboratory of Fujian Province, Fuzhou 350013, China
| | - Dingcheng Ye
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou 350013, China; (J.L.); (D.Y.)
- Livestock and Poultry Genetic Breeding Key Laboratory of Fujian Province, Fuzhou 350013, China
| | - Changkang Wang
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (X.W.); (C.X.); (S.W.); (Q.L.); (S.L.); (C.W.)
| | - Pingting Guo
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (X.W.); (C.X.); (S.W.); (Q.L.); (S.L.); (C.W.)
| |
Collapse
|
4
|
Gleeson PJ, Camara NOS, Launay P, Lehuen A, Monteiro RC. Immunoglobulin A Antibodies: From Protection to Harmful Roles. Immunol Rev 2024; 328:171-191. [PMID: 39578936 DOI: 10.1111/imr.13424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/15/2024] [Accepted: 11/07/2024] [Indexed: 11/24/2024]
Abstract
Immunoglobulin A (IgA) is the most abundantly produced antibody in humans. IgA is a unique class of immunoglobulin due to its multiple molecular forms, and a defining difference between the two subclasses: IgA1 has a long hinge-region that is heavily O-glycosylated, whereas the IgA2 hinge-region is shorter but resistant to bacterial proteases prevalent at mucosal sites. IgA is essential for immune homeostasis and education. Mucosal IgA plays a crucial role in maintaining the integrity of the mucosal barrier by immune exclusion of pathobionts while facilitating colonization with certain commensals; a large part of the gut microbiota is coated with IgA. In the circulation, monomeric IgA that has not been engaged by antigen plays a discrete role in dampening inflammatory responses. Protective and harmful roles of IgA have been studied over several decades, but a new understanding of the complex role of this immunoglobulin in health and disease has been provided by recent studies. Here, we discuss the physiological and pathological roles of IgA with a special focus on the gut, kidneys, and autoimmunity. We also discuss new IgA-based therapeutic approaches.
Collapse
Affiliation(s)
- Patrick J Gleeson
- Center for Research on Inflammation, Paris Cité University, Paris, France
- INSERM, Paris, France
- CNRS, Paris, France
- Inflamex Laboratory of Excellence, Paris, France
- Nephrology Department, Bichat Hospital, Paris, France
| | - Niels O S Camara
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Pierre Launay
- Center for Research on Inflammation, Paris Cité University, Paris, France
- INSERM, Paris, France
- CNRS, Paris, France
- Inflamex Laboratory of Excellence, Paris, France
| | - Agnès Lehuen
- Inflamex Laboratory of Excellence, Paris, France
- Cochin Institute, INSERM, CNRS, Paris Cité University, Paris, France
| | - Renato C Monteiro
- Center for Research on Inflammation, Paris Cité University, Paris, France
- INSERM, Paris, France
- CNRS, Paris, France
- Inflamex Laboratory of Excellence, Paris, France
| |
Collapse
|
5
|
Wang Y, Xiao J. Recent advances in the molecular understanding of immunoglobulin A. FEBS J 2024; 291:3597-3603. [PMID: 38329005 DOI: 10.1111/febs.17089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/11/2024] [Accepted: 01/31/2024] [Indexed: 02/09/2024]
Abstract
Immunoglobulin A (IgA) plays a crucial role in the human immune system, particularly in mucosal immunity. IgA antibodies that target the mucosal surface are made up of two to five IgA monomers linked together by the joining chain, forming polymeric molecules. These IgA polymers are transported across mucosal epithelial cells by the polymeric immunoglobulin receptor pIgR, resulting in the formation of secretory IgA (SIgA). This review aims to explore recent advancements in our molecular understanding of IgA, with a specific focus on SIgA, and the interaction between IgA and pathogen molecules.
Collapse
Affiliation(s)
- Yuxin Wang
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
| | - Junyu Xiao
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| |
Collapse
|
6
|
Ruocco V, Grünwald-Gruber C, Rad B, Tscheliessnig R, Hammel M, Strasser R. Effects of N-glycans on the structure of human IgA2. Front Mol Biosci 2024; 11:1390659. [PMID: 38645274 PMCID: PMC11026580 DOI: 10.3389/fmolb.2024.1390659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 03/22/2024] [Indexed: 04/23/2024] Open
Abstract
The transition of IgA antibodies into clinical development is crucial because they have the potential to create a new class of therapeutics with superior pathogen neutralization, cancer cell killing, and immunomodulation capacity compared to IgG. However, the biological role of IgA glycans in these processes needs to be better understood. This study provides a detailed biochemical, biophysical, and structural characterization of recombinant monomeric human IgA2, which varies in the amount/locations of attached glycans. Monomeric IgA2 antibodies were produced by removing the N-linked glycans in the CH1 and CH2 domains. The impact of glycans on oligomer formation, thermal stability, and receptor binding was evaluated. In addition, we performed a structural analysis of recombinant IgA2 in solution using Small Angle X-Ray Scattering (SAXS) to examine the effect of glycans on protein structure and flexibility. Our results indicate that the absence of glycans in the Fc tail region leads to higher-order aggregates. SAXS, combined with atomistic modeling, showed that the lack of glycans in the CH2 domain results in increased flexibility between the Fab and Fc domains and a different distribution of open and closed conformations in solution. When binding with the Fcα-receptor, the dissociation constant remains unaltered in the absence of glycans in the CH1 or CH2 domain, compared to the fully glycosylated protein. These results provide insights into N-glycans' function on IgA2, which could have important implications for developing more effective IgA-based therapeutics in the future.
Collapse
Affiliation(s)
- Valentina Ruocco
- Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Clemens Grünwald-Gruber
- Core Facility Mass Spectrometry, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Behzad Rad
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Rupert Tscheliessnig
- Division of Biophysics, Gottfried-Schatz-Research-Center, Medical University of Graz, Graz, Austria
| | - Michal Hammel
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Richard Strasser
- Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences, Vienna, Austria
| |
Collapse
|
7
|
Pan S, Manabe N, Ohno S, Komatsu S, Fujimura T, Yamaguchi Y. Each N-glycan on human IgA and J-chain uniquely affects oligomericity and stability. Biochim Biophys Acta Gen Subj 2024; 1868:130536. [PMID: 38070292 DOI: 10.1016/j.bbagen.2023.130536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 11/24/2023] [Accepted: 11/30/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND Immunoglobulin A (IgA) plays a pivotal role in various immune responses, especially that of mucosal immunity. IgA is usually assembled into dimers with the contribution of J-chains. There are two N-glycosylation sites in human IgA1-Fc and one in the J-chain. There is no consensus as yet on the functional role of the N-glycosylation. METHODS To gain a better understanding of their role, we designed a series of IgA1-Fc mutants, which were expressed in the absence or presence of the J-chain. RESULTS IgA1-Fc without the J-chain, was predominantly expressed as a monomer, and in its presence dimers and some polymers appeared. N263 (Fc Cα2), N459 (Fc tailpiece) and N49 (J-chain) were shown to be site-specifically modified with N-glycans by mass spectrometry analysis. Mutant IgA1-Fc N459Q failed to form a proper dimer in the presence of the J-chain, instead higher-order aggregates appeared. Fluorescence experiments suggest that the N459-glycans cover a hydrophobic surface at the Fc tailpiece that prevents other Fc molecules from approaching the dimeric IgA. A thermofluor assay revealed that the N-glycans at N263 (Fc) and N49 (J-chain) both contribute in different ways to the thermal stability of the Fc-J-chain complex. NMR analysis of 13C-labeled Fc suggests that the N459-glycan is relatively flexible while the N263-glycan is more rigid. CONCLUSIONS We conclude that the N459-glycan of IgA1-Fc is essential for dimer formation and prevention of higher-order aggregates while those at N263 (Fc) and N49 (J-chain) stabilize the Fc-J-chain complex. GENERAL SIGNIFICANCE Site-specific role for N-glycan in molecular assembly is addressed.
Collapse
Affiliation(s)
- Shunli Pan
- Division of Structural Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai 981-8558, Miyagi, Japan
| | - Noriyoshi Manabe
- Division of Structural Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai 981-8558, Miyagi, Japan
| | - Shiho Ohno
- Division of Structural Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai 981-8558, Miyagi, Japan
| | - Sachiko Komatsu
- Division of Bioanalytical Chemistry, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai 981-8558, Miyagi, Japan
| | - Tsutomu Fujimura
- Division of Bioanalytical Chemistry, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai 981-8558, Miyagi, Japan
| | - Yoshiki Yamaguchi
- Division of Structural Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai 981-8558, Miyagi, Japan.
| |
Collapse
|
8
|
Frerichs KA, Verkleij CPM, Mateos MV, Martin TG, Rodriguez C, Nooka A, Banerjee A, Chastain K, Perales-Puchalt A, Stephenson T, Uhlar C, Kobos R, van der Holt B, Kruyswijk S, Kuipers MT, Groen K, Vishwamitra D, Skerget S, Cortes-Selva D, Doyle M, Zaaijer HL, Zweegman S, Verona RI, van de Donk NWCJ. Teclistamab impairs humoral immunity in patients with heavily pretreated myeloma: importance of immunoglobulin supplementation. Blood Adv 2024; 8:194-206. [PMID: 38052042 PMCID: PMC10787247 DOI: 10.1182/bloodadvances.2023011658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/17/2023] [Accepted: 11/17/2023] [Indexed: 12/07/2023] Open
Abstract
ABSTRACT Teclistamab and other B-cell maturation antigen (BCMA)-targeting bispecific antibodies (BsAbs) have substantial activity in patients with heavily pretreated multiple myeloma (MM) but are associated with a high rate of infections. BCMA is also expressed on normal plasma cells and mature B cells, which are essential for the generation of a humoral immune response. The aim of this study was to improve the understanding of the impact of BCMA-targeting BsAbs on humoral immunity. The impact of teclistamab on polyclonal immunoglobulins and B cell counts was evaluated in patients with MM who received once-weekly teclistamab 1.5 mg/kg subcutaneously. Vaccination responses were assessed in a subset of patients. Teclistamabinduced rapid depletion of peripheral blood B cells in patients with MM and eliminated normal plasma cells in ex vivo assays. In addition, teclistamab reduced the levels of polyclonal immunoglobulins (immunoglobulin G [IgG], IgA, IgE, and IgM), without recovery over time while receiving teclistamab therapy. Furthermore, response to vaccines against Streptococcus pneumoniae, Haemophilus influenzae type B, and severe acute respiratory syndrome coronavirus 2 was severely impaired in patients treated with teclistamab compared with vaccination responses observed in patients with newly diagnosed MM or relapsed/refractory MM. Intravenous immunoglobulin (IVIG) use was associated with a significantly lower risk of serious infections among patients treated with teclistamab (cumulative incidence of infections at 6 months: 5.3% with IVIG vs 54.8% with observation only [P < .001]). In conclusion, our data show severe defects in humoral immunity induced by teclistamab, the impact of which can be mitigated by the use of immunoglobulin supplementation. This trial was registered at www.ClinicalTrials.gov as #NCT04557098.
Collapse
Affiliation(s)
- Kristine A Frerichs
- Department of Hematology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Christie P M Verkleij
- Department of Hematology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | | | | | | | - Ajay Nooka
- Winship Cancer Institute, Emory University, Atlanta, GA
| | | | | | | | | | | | - Rachel Kobos
- Janssen Research & Development, Spring House, PA
| | - Bronno van der Holt
- HOVON Foundation, Rotterdam, The Netherlands
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Sandy Kruyswijk
- Department of Hematology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Maria T Kuipers
- Department of Hematology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Kaz Groen
- Department of Hematology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | | | | | | | | | - Hans L Zaaijer
- Department of Medical Microbiology, Amsterdam UMC location, Academic Medical Center, Amsterdam, The Netherlands
| | - Sonja Zweegman
- Department of Hematology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | | | - Niels W C J van de Donk
- Department of Hematology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| |
Collapse
|
9
|
Shemesh S, Feldman R, Zagoory-Sharon O, Tzur Bitan D, Grossman-Giron A, Chen D, Maoz H, Bloch Y. Oxytocin as a transdiagnostic biomarker of well-being in severe mental illness during the Covid-19 pandemic. J Psychiatr Res 2024; 169:355-363. [PMID: 38101184 DOI: 10.1016/j.jpsychires.2023.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 09/04/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023]
Abstract
Individuals with severe mental illness (SMI) have been found to suffer a greater decline in psychological well-being compared to the general population in times of stress. The present study aimed to examine clinical and endocrine resilience factors of psychological well-being in SMI patients during the Covid-19 pandemic. METHODS After Covid-19 crisis outburst in Israel 112 participants, 69 outpatients, and 43 inpatients and day treatment patients were recruited. Outpatients signed an online informed consent and filled in questionnaires regarding their level of mental health symptoms (OQ-45), fear of Covid-19 (FCV), and psychological well-being (PWB). Inpatients answered the same questionnaires and in addition, went through a positive social interaction paradigm while providing three saliva samples to measure their s-IgA and oxytocin (OT) levels. RESULTS A strong negative correlation was found in the whole sample between reported mental health symptoms, fear of Covid-19, and well-being. Hierarchical regression did not find additional contribution of the fear of the pandemic in predicting well-being beyond the impact of symptomatology. For inpatients (N = 39) only, hierarchical regression found that oxytocin, but not s-IgA could explain 5% of the variance of well-being (R2 = 0.05) in individuals with SMI regardless of their mental health symptoms (R2 = 0.46) and their marital status (R2 = 0.21). CONCLUSIONS OT is suggested as a possible independent biological resilience factor of well-being in times of major stress among SMI patients. It is still unknown whether OT is a mediator that contributes to well-being or a biological marker that indicates the degree of beneficial social interactions.
Collapse
Affiliation(s)
| | | | | | - Dana Tzur Bitan
- Department of Community Mental Health, University of Haifa, Haifa, Israel
| | | | - Dror Chen
- Shalvata Mental Health Center, Hod- Hasharon, Israel; School of Medicine, Tel Aviv University, Ramat Aviv, Israel
| | - Hagai Maoz
- Shalvata Mental Health Center, Hod- Hasharon, Israel; School of Medicine, Tel Aviv University, Ramat Aviv, Israel
| | - Yuval Bloch
- School of Medicine, Tel Aviv University, Ramat Aviv, Israel.
| |
Collapse
|
10
|
Mane V, Mehta R, Alvarez N, Sharma V, Park S, Fox A, DeCarlo C, Yang X, Perlin DS, Powell RLR. In Vivo Antiviral Efficacy of LCTG-002, a Pooled, Purified Human Milk Secretory IgA product, Against SARS-CoV-2 in a Murine Model of COVID-19. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.25.554813. [PMID: 37693438 PMCID: PMC10491103 DOI: 10.1101/2023.08.25.554813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Immunoglobulin A (IgA) is the most abundant antibody (Ab) in human mucosal compartments including the respiratory tract, with the secretory form of IgA (sIgA) being dominant and uniquely stable in these environments. sIgA is naturally found in human milk, which could be considered a global resource for this biologic, justifying the development of human milk sIgA as a dedicated airway therapeutic for respiratory infections such as SARS-CoV-2. In the present study, methods were therefore developed to efficiently extract human milk sIgA from donors who were either immunologically naïve to SARS-CoV-2 (pooled as a control IgA) or had recovered from a PCR-confirmed SARS-CoV-2 infection that elicited high-titer anti-SARS-CoV-2 Spike sIgA Abs in their milk (pooled together to make LCTG-002). Mass spectrometry determined that proteins with a relative abundance of 1.0% or greater were all associated with sIgA. None of the proteins exhibited statistically significant differences between batches. Western blot demonstrated all batches consisted predominantly of sIgA. Compared to control IgA, LCTG-002 demonstrated significantly higher binding to Spike, and was also capable of blocking the Spike - ACE2 interaction in vitro with 6.3x greater potency compared to control IgA (58% inhibition at ∼240ug/mL). LCTG-002 was then tested in vivo for its capacity to reduce viral burden in the lungs of K18+hACE2 transgenic mice inoculated with SARS-CoV-2. LCTG-002 was demonstrated to significantly reduce SARS-CoV-2 titers in the lungs compared to control IgA when administered at either 250ug/day or 1 mg/day, as measured by TCID50, plaque forming units (PFU), and qRT-PCR, with a maximum reduction of 4.9 logs. This innovative study demonstrates that LCTG-002 is highly pure, efficacious, and well tolerated in vivo, supporting further development of milk-derived, polyclonal sIgA therapeutics against SARS-CoV-2 and other mucosal infections.
Collapse
|
11
|
Palwankar P, Jain S, Pandey R, Mahesh S. IgA Levels among Type 2 Diabetic and Non-Diabetic Patients with Periodontitis: A Prospective Clinical Study. Eur J Dent 2023; 17:823-827. [PMID: 36167319 PMCID: PMC10569843 DOI: 10.1055/s-0042-1755616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
Abstract
OBJECTIVES To estimate gingival crevicular immunoglobulin A(IgA) using enzyme-linked immunosorbent assay (ELISA) among type II diabetic patients with periodontitis. MATERIALS AND METHODS A non-randomized study was done of 40 periodontitis subjects with a mean age of 50 years and were recruited into two groups, Group A (Type II controlled diabetics with HbA1c < 7%) and Group B (non-diabetics with HbA1c between 4 and 6%). Both the groups underwent nonsurgical periodontal therapy (NSPT). The clinical parameters were recorded at baseline, 1, and 3 months. GCF sample was collected for the estimation of crevicular IgA at baseline and at 3 months. STATISTICAL ANALYSIS Results were analyzed using parametric tests paired t-test and Student's t-test for every assessment point. The level of significance was set at p < 0.05. RESULTS Difference in IgA levels and clinical parameters was seen between diabetic and non-diabetic groups, which was statistically significant. CONCLUSION Changes in crevicular IgA levels in patients with diabetic periodontitis can be used as a novel biomarker in assessing the inflammatory status.
Collapse
Affiliation(s)
- Pooja Palwankar
- Department of Periodontology, Manav Rachna Dental College FDS, MRIIRS Faridabad, Haryana, India
| | - Saumya Jain
- Department of Periodontology, Manav Rachna Dental College FDS, MRIIRS Faridabad, Haryana, India
| | - Ruchi Pandey
- Department of Periodontology, Manav Rachna Dental College FDS, MRIIRS Faridabad, Haryana, India
| | - Shakila Mahesh
- Department of Microbiology, Manav Rachna Dental College FDS, MRIIRS Faridabad, Haryana, India
| |
Collapse
|
12
|
Bohländer F. A new hope? Possibilities of therapeutic IgA antibodies in the treatment of inflammatory lung diseases. Front Immunol 2023; 14:1127339. [PMID: 37051237 PMCID: PMC10083398 DOI: 10.3389/fimmu.2023.1127339] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/14/2023] [Indexed: 03/29/2023] Open
Abstract
Inflammatory lung diseases represent a persistent burden for patients and the global healthcare system. The combination of high morbidity, (partially) high mortality and limited innovations in the last decades, have resulted in a great demand for new therapeutics. Are therapeutic IgA antibodies possibly a new hope in the treatment of inflammatory lung diseases? Current research increasingly unravels the elementary functions of IgA as protector against infections and as modulator of overwhelming inflammation. With a focus on IgA, this review describes the pathological alterations in mucosal immunity and how they contribute to chronic inflammation in the most common inflammatory lung diseases. The current knowledge of IgA functions in the circulation, and particularly in the respiratory mucosa, are summarized. The interplay between neutrophils and IgA seems to be key in control of inflammation. In addition, the hurdles and benefits of therapeutic IgA antibodies, as well as the currently known clinically used IgA preparations are described. The data highlighted here, together with upcoming research strategies aiming at circumventing the current pitfalls in IgA research may pave the way for this promising antibody class in the application of inflammatory lung diseases.
Collapse
Affiliation(s)
- Fabian Bohländer
- Department of Translational Research, Biotest AG, Dreieich, Germany
| |
Collapse
|
13
|
Hamdan F, Cerullo V. Cancer immunotherapies: A hope for the uncurable? FRONTIERS IN MOLECULAR MEDICINE 2023; 3:1140977. [PMID: 39086690 PMCID: PMC11285639 DOI: 10.3389/fmmed.2023.1140977] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 02/07/2023] [Indexed: 08/02/2024]
Abstract
The use of cancer immunotherapies is not novel but has been used over the decades in the clinic. Only recently have we found the true potential of stimulating an anti-tumor response after the breakthrough of checkpoint inhibitors. Cancer immunotherapies have become the first line treatment for many malignancies at various stages. Nevertheless, the clinical results in terms of overall survival and progression free survival were not as anticipated. Majority of cancer patients do not respond to immunotherapies and the reasons differ. Hence, further improvements for cancer immunotherapies are crucially needed. In the review, we will discuss various forms of cancer immunotherapies that are being tested or already in the clinic. Moreover, we also highlight future directions to improve such therapies.
Collapse
Affiliation(s)
- Firas Hamdan
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- TRIMM, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- Drug Delivery, Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Vincenzo Cerullo
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- TRIMM, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- Drug Delivery, Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
- Department of Molecular Medicine and Medical Biotechnology and CEINGE, Naples University Federico II, Naples, Italy
| |
Collapse
|
14
|
Sewnath CA, Behrens LM, van Egmond M. Targeting myeloid cells with bispecific antibodies as novel immunotherapies of cancer. Expert Opin Biol Ther 2022; 22:983-995. [PMID: 35854649 DOI: 10.1080/14712598.2022.2098675] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Most bispecific antibody (BsAb) therapies focus on stimulating the adaptive immune system, in particular T cells, to promote tumor cell killing. Another method to promote tumor eradication is through the engagement of myeloid cells, including macrophages and neutrophils, which are abundantly present and possess intrinsic cytotoxic mechanisms for tumor cell killing, making them interesting effector cells to recruit for BsAb therapy. AREAS COVERED In this review, we describe the evolving knowledge of the role of macrophages and neutrophils in cancer in scientific literature. Moreover, we address the BsAbs that have been developed over the years to recruit these cell types as effector cells in immunotherapy of cancer. This includes the discussion of BsAbs that target Fc receptors (i.e. FcγR and FcαRI) to induce antibody-dependent cellular phagocytosis (ADCP) by macrophages or trogoptosis via neutrophils, as well as BsAbs that interfere with checkpoint inhibition, including the SIRPα-CD47 pathway. EXPERT OPINION Elucidating the complexity of macrophage and neutrophil heterogeneity in cancer may help to specifically enlist the cytotoxic ability of these cells through targeting Fc receptors and checkpoint pathways, which may further enhance anti-cancer immunity.
Collapse
Affiliation(s)
- Celine An Sewnath
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam De Boelelaan, Amsterdam, The Netherlands.,Cancer Biology and Immunology Program, Cancer Centre Amsterdam, Amsterdam, The Netherlands.,Cancer Immunology Program, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Leonie M Behrens
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam De Boelelaan, Amsterdam, The Netherlands.,Cancer Biology and Immunology Program, Cancer Centre Amsterdam, Amsterdam, The Netherlands.,Cancer Immunology Program, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Marjolein van Egmond
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam De Boelelaan, Amsterdam, The Netherlands.,Cancer Biology and Immunology Program, Cancer Centre Amsterdam, Amsterdam, The Netherlands.,Cancer Immunology Program, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands.,Department of Surgery, Amsterdam UMC, Vrije Universiteit Amsterdam De Boelelaan, Amsterdam, The Netherlands
| |
Collapse
|
15
|
Behrens LM, van den Berg TK, van Egmond M. Targeting the CD47-SIRPα Innate Immune Checkpoint to Potentiate Antibody Therapy in Cancer by Neutrophils. Cancers (Basel) 2022; 14:cancers14143366. [PMID: 35884427 PMCID: PMC9319280 DOI: 10.3390/cancers14143366] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Immunotherapy aims to engage various immune cells in the elimination of cancer cells. Neutrophils are the most abundant leukocytes in the circulation and have unique mechanisms by which they can kill cancer cells opsonized by antibodies. However, neutrophil effector functions are limited by the inhibitory receptor SIRPα, when it interacts with CD47. The CD47 protein is expressed on all cells in the body and acts as a ‘don’t eat me’ signal to prevent tissue damage. Cancer cells can express high levels of CD47 to circumvent tumor elimination. Thus, blocking the interaction between CD47 and SIRPα may enhance anti-tumor effects by neutrophils in the presence of tumor-targeting monoclonal antibodies. In this review, we discuss CD47-SIRPα as an innate immune checkpoint on neutrophils and explore the preliminary results of clinical trials using CD47-SIRPα blocking agents. Abstract In the past 25 years, a considerable number of therapeutic monoclonal antibodies (mAb) against a variety of tumor-associated antigens (TAA) have become available for the targeted treatment of hematologic and solid cancers. Such antibodies opsonize cancer cells and can trigger cytotoxic responses mediated by Fc-receptor expressing immune cells in the tumor microenvironment (TME). Although frequently ignored, neutrophils, which are abundantly present in the circulation and many cancers, have demonstrated to constitute bona fide effector cells for antibody-mediated tumor elimination in vivo. It has now also been established that neutrophils exert a unique mechanism of cytotoxicity towards antibody-opsonized tumor cells, known as trogoptosis, which involves Fc-receptor (FcR)-mediated trogocytosis of cancer cell plasma membrane leading to a lytic/necrotic type of cell death. However, neutrophils prominently express the myeloid inhibitory receptor SIRPα, which upon interaction with the ‘don’t eat me’ signal CD47 on cancer cells, limits cytotoxicity, forming a mechanism of resistance towards anti-cancer antibody therapeutics. In fact, tumor cells often overexpress CD47, thereby even more strongly restricting neutrophil-mediated tumor killing. Blocking the CD47-SIRPα interaction may therefore potentiate neutrophil-mediated antibody-dependent cellular cytotoxicity (ADCC) towards cancer cells, and various inhibitors of the CD47-SIRPα axis are now in clinical studies. Here, we review the role of neutrophils in antibody therapy in cancer and their regulation by the CD47-SIRPα innate immune checkpoint. Moreover, initial results of CD47-SIRPα blockade in clinical trials are discussed.
Collapse
Affiliation(s)
- Leonie M. Behrens
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (T.K.v.d.B.); (M.v.E.)
- Cancer Center Amsterdam, Cancer Biology and Immunology Program, 1081 HV Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology Program, 1081 HV Amsterdam, The Netherlands
- Correspondence:
| | - Timo K. van den Berg
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (T.K.v.d.B.); (M.v.E.)
- Byondis B.V., 6545 CM Nijmegen, The Netherlands
| | - Marjolein van Egmond
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (T.K.v.d.B.); (M.v.E.)
- Cancer Center Amsterdam, Cancer Biology and Immunology Program, 1081 HV Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology Program, 1081 HV Amsterdam, The Netherlands
- Department of Surgery, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
16
|
Chai J, Sun Z, Zhang S, Zhou Q, Xu J. An indirect approach for reference intervals establishment of immunoglobulin A, G and M: the combination of laboratory database and statistics. Scandinavian Journal of Clinical and Laboratory Investigation 2022; 82:311-322. [PMID: 35791063 DOI: 10.1080/00365513.2022.2092897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Immunoglobulins are affected by sex, age and region, so it is necessary to establish suitable reference intervals (RIs) for clinical diagnosis. Various statistical methods were used to calculate RIs, but there has been a lack of comparison among the methods. Research based on immunoglobulin RIs establishment with various methods would provide a methodological basis for further research. A total of 16,525 individuals were enrolled in the study. Individuals were selected in the medical examination center of First Hospital of Jilin University from 2014 to 2020. The lambda-mu-sigma (LMS) method was performed to evaluate the dynamic changes in analytes. RIs were calculated by parametric, non-parametric, Hoffman method and Bhattacharya method. Sex and age partitions were found for immunoglobulins G and immunoglobulin M. The levels of IgM showed no difference with age in males, but showed differences after 50 years of age in females. Circulating immunoglobulin A concentrations showed an increasing trend with age, and immunoglobulin M showed a fluctuating trend with age. Obvious difference (>5%) was commonly found among the four methods, however, the RIs established by the four methods all passed the verification with a high passing rate. Sex and age differences should be considered for immunoglobulins G and immunoglobulin M in clinical practice. The feasibility of the four indirect methods was proven, which would provides a methodological reference for further studies and benefit the application of clinical data.
Collapse
Affiliation(s)
- Jiatong Chai
- Department of Laboratory Medicine, The First Hospital of Jilin University, Changchun, China
| | - Zeyu Sun
- Department of Laboratory Medicine, The First Hospital of Jilin University, Changchun, China
| | - Shunli Zhang
- Department of Clinical Laboratory, Beijing Chaoyang Hospital, The Third Clinical Medical College of Capital Medical University, Beijing Center for Clinical Laboratories, Beijing, China
| | - Qi Zhou
- The Department of Pediatrics at the First Hospital of Jilin University, Changchun, China
| | - Jiancheng Xu
- Department of Laboratory Medicine, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
17
|
Ling WL, Su CTT, Lua WH, Yeo JY, Poh JJ, Ng YL, Wipat A, Gan SKE. Variable-heavy (VH) families influencing IgA1&2 engagement to the antigen, FcαRI and superantigen proteins G, A, and L. Sci Rep 2022; 12:6510. [PMID: 35444201 PMCID: PMC9020155 DOI: 10.1038/s41598-022-10388-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 04/07/2022] [Indexed: 12/18/2022] Open
Abstract
Interest in IgA as an alternative antibody format has increased over the years with much remaining to be investigated in relation to interactions with immune cells. Considering the recent whole antibody investigations showing significant distal effects between the variable (V) and constant (C)- regions that can be mitigated by the hinge regions of both human IgA subtypes A1 and A2, we performed an in-depth mechanistic investigation using a panel of 28 IgA1s and A2s of both Trastuzumab and Pertuzumab models. FcαRI binding were found to be mitigated by the differing glycosylation patterns in IgA1 and 2 with contributions from the CDRs. On their interactions with antigen-Her2 and superantigens PpL, SpG and SpA, PpL was found to sterically hinder Her2 antigen binding with unexpected findings of IgAs binding SpG at the CH2-3 region alongside SpA interacting with IgAs at the CH1. Although the VH3 framework (FWR) is commonly used in CDR grafting, we found the VH1 framework (FWR) to be a possible alternative when grafting IgA1 and 2 owing to its stronger binding to antigen Her2 and weaker interactions to superantigen Protein L and A. These findings lay the foundation to understanding the interactions between IgAs and microbial superantigens, and also guide the engineering of IgAs for future antibody applications and targeting of superantigen-producing microbes.
Collapse
Affiliation(s)
- Wei-Li Ling
- Antibody & Product Development Lab, Experimental Drug Development Centre, Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.,Newcastle University Singapore, Singapore, Singapore
| | - Chinh Tran-To Su
- Antibody & Product Development Lab, Experimental Drug Development Centre, Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Wai-Heng Lua
- Antibody & Product Development Lab, Experimental Drug Development Centre, Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Joshua Yi Yeo
- Antibody & Product Development Lab, Experimental Drug Development Centre, Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Jun-Jie Poh
- Antibody & Product Development Lab, Experimental Drug Development Centre, Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Yuen-Ling Ng
- Newcastle University Singapore, Singapore, Singapore
| | - Anil Wipat
- School of Computing, Newcastle University, Newcastle upon Tyne, UK
| | - Samuel Ken-En Gan
- Antibody & Product Development Lab, Experimental Drug Development Centre, Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore. .,James Cook University, Singapore, Singapore. .,Zhejiang Bioinformatics International Science and Technology Cooperation Center, Wenzhou-Kean University, Wenzhou, Zhejiang Province, China. .,Wenzhou Municipal Key Lab of Applied Biomedical and Biopharmaceutical Informatics, Wenzhou-Kean University, Wenzhou, Zhejiang Province, China.
| |
Collapse
|
18
|
de Fays C, Carlier FM, Gohy S, Pilette C. Secretory Immunoglobulin A Immunity in Chronic Obstructive Respiratory Diseases. Cells 2022; 11:1324. [PMID: 35456002 PMCID: PMC9027823 DOI: 10.3390/cells11081324] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/01/2022] [Accepted: 04/08/2022] [Indexed: 02/01/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD), asthma and cystic fibrosis (CF) are distinct respiratory diseases that share features such as the obstruction of small airways and disease flare-ups that are called exacerbations and are often caused by infections. Along the airway epithelium, immunoglobulin (Ig) A contributes to first line mucosal protection against inhaled particles and pathogens. Dimeric IgA produced by mucosal plasma cells is transported towards the apical pole of airway epithelial cells by the polymeric Ig receptor (pIgR), where it is released as secretory IgA. Secretory IgA mediates immune exclusion and promotes the clearance of pathogens from the airway surface by inhibiting their adherence to the epithelium. In this review, we summarize the current knowledge regarding alterations of the IgA/pIgR system observed in those major obstructive airway diseases and discuss their implication for disease pathogenesis.
Collapse
Affiliation(s)
- Charlotte de Fays
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium; (C.d.F.); (F.M.C.); (S.G.)
| | - François M. Carlier
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium; (C.d.F.); (F.M.C.); (S.G.)
- Department of Pneumology, CHU UCL Namur, Site Mont-Godinne, 5530 Yvoir, Belgium
- Lung Transplant Centre, CHU UCL Namur, Site Mont-Godinne, 5530 Yvoir, Belgium
| | - Sophie Gohy
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium; (C.d.F.); (F.M.C.); (S.G.)
- Department of Pneumology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
- Cystic Fibrosis Reference Centre, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Charles Pilette
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium; (C.d.F.); (F.M.C.); (S.G.)
- Department of Pneumology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| |
Collapse
|
19
|
Gruijs M, Sewnath CAN, Egmond MV. Therapeutic exploitation of neutrophils to fight cancer. Semin Immunol 2021; 57:101581. [PMID: 34922817 DOI: 10.1016/j.smim.2021.101581] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/22/2021] [Accepted: 12/09/2021] [Indexed: 12/18/2022]
Abstract
Antibody-based immunotherapy is a promising strategy in cancer treatment. Antibodies can directly inhibit tumor growth, induce complement-dependent cytotoxicity and induce Fc receptor-mediated elimination of tumor cells by macrophages and natural killer cells. Until now, however, neutrophils have been largely overlooked as potential effector cells, even though they are the most abundant type of immune cells in the circulation. Neutrophils display heterogeneity, especially in the context of cancer. Therefore, their role in cancer is debated. Nevertheless, neutrophils possess natural anti-tumor properties and appropriate stimulation, i.e. specific targeting via antibody therapy, induces potent tumor cell killing, especially via targeting of the immunoglobulin A Fc receptor (FcαRI, CD89). In this review we address the mechanisms of tumor cell killing by neutrophils and the role of neutrophils in induction of anti-tumor immunity. Moreover, possibilities for therapeutic targeting are discussed.
Collapse
Affiliation(s)
- Mandy Gruijs
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam - Amsterdam Institute for Infection and Immunity, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Celine A N Sewnath
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam - Amsterdam Institute for Infection and Immunity, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Marjolein van Egmond
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam - Amsterdam Institute for Infection and Immunity, De Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Surgery, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands.
| |
Collapse
|
20
|
Bohländer F, Weißmüller S, Riehl D, Gutscher M, Schüttrumpf J, Faust S. The Functional Role of IgA in the IgM/IgA-Enriched Immunoglobulin Preparation Trimodulin. Biomedicines 2021; 9:1828. [PMID: 34944644 PMCID: PMC8698729 DOI: 10.3390/biomedicines9121828] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 11/16/2022] Open
Abstract
In comparison to human immunoglobulin (Ig) G, antibodies of IgA class are not well investigated. In line with this, the functional role of the IgA component in IgM/IgA-enriched immunoglobulin preparations is also largely unknown. In recent years, powerful anti-pathogenic and immunomodulatory properties of human serum IgA especially on neutrophil function were unraveled. Therefore, the aim of our work is to investigate functional aspects of the trimodulin IgA component, a new plasma-derived polyvalent immunoglobulin preparation containing ~56% IgG, ~23% IgM and ~21% IgA. The functional role of IgA was investigated by analyzing the interaction of IgA with FcαRI, comparing trimodulin with standard intravenous IgG (IVIG) preparation and investigating Fc receptor (FcR)-dependent functions by excluding IgM-mediated effects. Trimodulin demonstrated potent immunomodulatory, as well as anti-pathogenic effects in our neutrophil model (neutrophil-like HL-60 cells). The IgA component of trimodulin was shown to induce a strong FcαRI-dependent inhibitory immunoreceptor tyrosine-based activation motif (ITAMi) signaling, counteract lipopolysaccharide-induced inflammation and mediate phagocytosis of Staphylococcus aureus. The fine-tuned balance between immunomodulatory and anti-pathogenic effects of trimodulin were shown to be dose-dependent. Summarized, our data demonstrate the functional role of IgA in trimodulin, highlighting the importance of this immunoglobulin class in immunoglobulin therapy.
Collapse
Affiliation(s)
- Fabian Bohländer
- Department of Analytical Development and Validation, Biotest AG, Landsteinerstraße 5, 63303 Dreieich, Germany; (F.B.); (D.R.); (M.G.)
| | - Sabrina Weißmüller
- Department of Translational Research, Biotest AG, Landsteinerstraße 5, 63303 Dreieich, Germany;
| | - Dennis Riehl
- Department of Analytical Development and Validation, Biotest AG, Landsteinerstraße 5, 63303 Dreieich, Germany; (F.B.); (D.R.); (M.G.)
| | - Marcus Gutscher
- Department of Analytical Development and Validation, Biotest AG, Landsteinerstraße 5, 63303 Dreieich, Germany; (F.B.); (D.R.); (M.G.)
| | - Jörg Schüttrumpf
- Corporate R&D, Biotest AG, Landsteinerstraße 5, 63303 Dreieich, Germany;
| | - Stefanie Faust
- Department of Analytical Development and Validation, Biotest AG, Landsteinerstraße 5, 63303 Dreieich, Germany; (F.B.); (D.R.); (M.G.)
| |
Collapse
|
21
|
Gorshkova EN, Pashova S, Vasilenko EA, Tchurina TS, Razzorenova EA, Starkina OV, Dimitrova P, Pashov A, Vassilev TL. Induced Polyspecificity of Human Secretory Immunoglobulin A Antibodies: Is It Possible to Improve Their Ability to Bind Pathogens? Pharmacology 2021; 107:341-350. [PMID: 34864734 DOI: 10.1159/000520343] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 10/16/2021] [Indexed: 11/19/2022]
Abstract
INTRODUCTION As has been shown previously, various protein-modifying agents can change the antigen-binding properties of immunoglobulins. However, induced polyspecificity of human secretory immunoglobulin A (sIgA) has not been previously characterized in detail. METHODS In the present study, human secretory immunoglobulin A (IgA) was exposed to buffers with acidic pH, to free heme, or to pro-oxidative ferrous ions, and the antigen-binding behavior of the native and modified IgA to viral and bacterial antigens was compared using Western blotting and enzyme-linked immunosorbent assay. The ability of these agents to modulate the antigen-binding properties of human sIgA toward a wide range of pathogen peptides was investigated using an epitope microarray. RESULTS We have shown that acidic pH, heme, and pro-oxidative ferrous ions influenced the binding of secretory IgA in opposite directions (either increasing or decreasing); however, the strongest effect was observed when using buffers with low pH. This fraction had the highest number of affected reactivities; most of them were increased and most of the new ones were toward common pathogens. CONCLUSIONS Thus, it was shown that all investigated treatments can alter to some degree the antigen-binding of secretory IgA, but acidic pH has the most potentially beneficial effect by increasing binding to a largest number of common pathogens' antigens.
Collapse
Affiliation(s)
- Ekaterina N Gorshkova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russian Federation
| | - Shina Pashova
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Ekaterina A Vasilenko
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russian Federation
| | - Tatiana S Tchurina
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russian Federation
| | - Elizaveta A Razzorenova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russian Federation
| | - Olga V Starkina
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russian Federation
| | - Petya Dimitrova
- Department of Immunology, Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Anastas Pashov
- Department of Immunology, Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Tchavdar Lubenov Vassilev
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russian Federation.,Department of Immunology, Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| |
Collapse
|
22
|
Comparison of Overall Immunity Levels among Workers at Grape Orchard, Rose Greenhouse, and Open-Field Onion Farm. Saf Health Work 2021; 13:248-254. [PMID: 35664906 PMCID: PMC9142355 DOI: 10.1016/j.shaw.2021.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 11/03/2021] [Accepted: 12/17/2021] [Indexed: 11/23/2022] Open
Abstract
Background Methods Results Conclusion
Collapse
|
23
|
Yamaki K, Terashi M, Yamamoto S, Fujiwara R, Inoue JI, Shimizu K, Yanagita S, Doi Y, Kimura KI, Kotani K, Sugihara M, Koyama Y. Immunoglobulin a suppresses B cell receptor-mediated activation of mouse B cells with differential inhibition of signaling molecules. Immunopharmacol Immunotoxicol 2021; 44:76-86. [PMID: 34844505 DOI: 10.1080/08923973.2021.2006216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
CONTEXT We previously reported that monoclonal mouse immunoglobulin (Ig) A, OA-4, attenuates sensitization in mice by suppressing B cell activation. OBJECTIVE Here, it is demonstrated for the first time that mouse IgA inhibits mouse B cell activation in vitro under natural conditions (i.e. in the absence of chemical, physical, and genetic modifications of IgA and B cells). MATERIALS AND METHODS Mouse splenocytes were stimulated with anti-B cell receptor (BCR) antibody or lipopolysaccharide (LPS) in the presence or absence of OA-4. Splenic B cell proliferation and the activation of several intracellular signaling molecules were measured. RESULTS Anti-BCR antibody-induced proliferation was markedly inhibited by OA-4 or the commercially available mouse IgA S107, whereas LPS-induced proliferation was weakly attenuated by a high concentration of OA-4. Moreover, OA-4 markedly decreased the anti-BCR antibody-induced phosphorylation of p44/42 mitogen-activated protein kinase (ERK) and CD22 and decreased phosphorylated phospholipase (PLC) γ2 and intracellular Ca2+ levels moderately, whereas protein kinase B (Akt) phosphorylation was not affected by OA-4. The MAPK/ERK kinase-ERK and phosphoinositide 3-kinase-Akt pathways were found to play a role in the proliferation of splenocytes induced by anti-BCR antibody based on experiments with their inhibitors. In contrast to that in splenic B cells, ERK phosphorylation induced by anti-BCR antibody in A20 cells was not inhibited by OA-4. The modulatory effects of IgA were different among the cell types and signaling pathways. CONCLUSION IgA is a potential immunoregulatory drug utilizing new mechanisms that affect splenic B cells but not A20 lymphomas.
Collapse
Affiliation(s)
- Kouya Yamaki
- Laboratory of Pharmacology, Kobe Pharmaceutical University, Kobe, Japan
| | - Masato Terashi
- Laboratory of Pharmacology, Kobe Pharmaceutical University, Kobe, Japan
| | - Saori Yamamoto
- Laboratory of Pharmacology, Kobe Pharmaceutical University, Kobe, Japan
| | - Rei Fujiwara
- Laboratory of Pharmacology, Kobe Pharmaceutical University, Kobe, Japan
| | - Jun-Ichi Inoue
- Laboratory of Pharmacology, Kobe Pharmaceutical University, Kobe, Japan
| | - Kishi Shimizu
- Laboratory of Pharmacology, Kobe Pharmaceutical University, Kobe, Japan
| | - Sakura Yanagita
- Laboratory of Pharmacology, Kobe Pharmaceutical University, Kobe, Japan
| | - Yuma Doi
- Laboratory of Pharmacology, Kobe Pharmaceutical University, Kobe, Japan
| | - Kei-Ichiro Kimura
- Laboratory of Pharmacology, Kobe Pharmaceutical University, Kobe, Japan
| | - Kayo Kotani
- Laboratory of Pharmacology, Kobe Pharmaceutical University, Kobe, Japan
| | - Mai Sugihara
- Laboratory of Pharmacology, Kobe Pharmaceutical University, Kobe, Japan
| | - Yutaka Koyama
- Laboratory of Pharmacology, Kobe Pharmaceutical University, Kobe, Japan
| |
Collapse
|
24
|
3D Structures of IgA, IgM, and Components. Int J Mol Sci 2021; 22:ijms222312776. [PMID: 34884580 PMCID: PMC8657937 DOI: 10.3390/ijms222312776] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 11/20/2021] [Accepted: 11/23/2021] [Indexed: 12/02/2022] Open
Abstract
Immunoglobulin G (IgG) is currently the most studied immunoglobin class and is frequently used in antibody therapeutics in which its beneficial effector functions are exploited. IgG is composed of two heavy chains and two light chains, forming the basic antibody monomeric unit. In contrast, immunoglobulin A (IgA) and immunoglobulin M (IgM) are usually assembled into dimers or pentamers with the contribution of joining (J)-chains, which bind to the secretory component (SC) of the polymeric Ig receptor (pIgR) and are transported to the mucosal surface. IgA and IgM play a pivotal role in various immune responses, especially in mucosal immunity. Due to their structural complexity, 3D structural study of these molecules at atomic scale has been slow. With the emergence of cryo-EM and X-ray crystallographic techniques and the growing interest in the structure-function relationships of IgA and IgM, atomic-scale structural information on IgA-Fc and IgM-Fc has been accumulating. Here, we examine the 3D structures of IgA and IgM, including the J-chain and SC. Disulfide bridging and N-glycosylation on these molecules are also summarized. With the increasing information of structure–function relationships, IgA- and IgM-based monoclonal antibodies will be an effective option in the therapeutic field.
Collapse
|
25
|
Shinkura R. Therapeutic immunoglobulin A antibody for dysbiosis-related diseases. Int Immunol 2021; 33:787-790. [PMID: 34492105 PMCID: PMC8499904 DOI: 10.1093/intimm/dxab066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/07/2021] [Indexed: 11/12/2022] Open
Abstract
Dysbiosis is alterations in the microbial composition compared with a healthy microbiota and often features a reduction in gut microbial diversity and a change in microbial taxa. Dysbiosis, especially in the gut, has also been proposed to play a crucial role in the pathogenesis of a wide variety of diseases, including inflammatory bowel disease, colorectal cancer, cardiovascular disease, obesity, diabetes and multiple sclerosis. A body of evidence has shown that intestinal polymeric immunoglobulin A (IgA) antibodies are important to regulate the gut microbiota as well as to exclude pathogenic bacteria or viral infection such as influenza and SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) at mucosal sites. Since the 1970s, trials for oral administration of therapeutic IgA or IgG have been performed mainly to treat infectious enteritis caused by pathogenic Escherichia coli or Clostridium difficile. However, few of them have been successfully developed for clinical application up to now. In addition to the protective function against intestinal pathogens, IgA is well known to modulate the gut commensal microbiota leading to symbiosis. Nevertheless, the development of therapeutic IgA drugs to treat dysbiosis is not progressing. In this review, the advantages of therapeutic IgA antibodies and the problems for their development will be discussed.
Collapse
Affiliation(s)
- Reiko Shinkura
- Laboratory of Immunology and Infection Control, Institute for Quantitative Biosciences, University of Tokyo, Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
- Collaborative Research Institute for Innovative Microbiology, University of Tokyo, 1‑1‑1 Yayoi, Bunkyo‑ku, Tokyo 113‑0032, Japan
- Core Research for Evolutional Science and Technology, Japan Agency for Medical Research and Development, Tokyo 100-0004, Japan
| |
Collapse
|
26
|
IgA Vasculitis: Etiology, Treatment, Biomarkers and Epigenetic Changes. Int J Mol Sci 2021; 22:ijms22147538. [PMID: 34299162 PMCID: PMC8307949 DOI: 10.3390/ijms22147538] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 07/12/2021] [Indexed: 02/06/2023] Open
Abstract
IgA, previously called Henoch-Schönlein vasculitis, is an essential immune component that drives the host immune response to the external environment. As IgA has the unique characteristic of a flexible response to broad types of microorganisms, it sometimes causes an autoreactive response in the host human body. IgA vasculitis and related organ dysfunction are representative IgA-mediated autoimmune diseases; bacterial and viral infections often trigger IgA vasculitis. Recent drug developments and the presence of COVID-19 have revealed that these agents can also trigger IgA vasculitis. These findings provide a novel understanding of the pathogenesis of IgA vasculitis. In this review, we focus on the characteristics of IgA and symptoms of IgA vasculitis and other organ dysfunction. We also mention the therapeutic approach, biomarkers, novel triggers for IgA vasculitis, and epigenetic modifications in patients with IgA vasculitis.
Collapse
|
27
|
Bohländer F, Riehl D, Weißmüller S, Gutscher M, Schüttrumpf J, Faust S. Immunomodulation: Immunoglobulin Preparations Suppress Hyperinflammation in a COVID-19 Model via FcγRIIA and FcαRI. Front Immunol 2021; 12:700429. [PMID: 34177967 PMCID: PMC8223875 DOI: 10.3389/fimmu.2021.700429] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 05/21/2021] [Indexed: 12/13/2022] Open
Abstract
The rapid spread of SARS-CoV-2 has induced a global pandemic. Severe forms of COVID-19 are characterized by dysregulated immune response and "cytokine storm". The role of IgG and IgM antibodies in COVID-19 pathology is reasonably well studied, whereas IgA is neglected. To improve clinical outcome of patients, immune modulatory drugs appear to be beneficial. Such drugs include intravenous immunoglobulin preparations, which were successfully tested in severe COVID-19 patients. Here we established a versatile in vitro model to study inflammatory as well as anti-inflammatory processes by therapeutic human immunoglobulins. We dissect the inflammatory activation on neutrophil-like HL60 cells, using an immune complex consisting of latex beads coated with spike protein of SARS-CoV-2 and opsonized with specific immunoglobulins from convalescent plasma. Our data clarifies the role of Fc-receptor-dependent phagocytosis via IgA-FcαRI and IgG-FcγR for COVID-19 disease followed by cytokine release. We show that COVID-19 associated inflammation could be reduced by addition of human immunoglobulin preparations (IVIG and trimodulin), while trimodulin elicits stronger immune modulation by more powerful ITAMi signaling. Besides IgG, the IgA component of trimodulin in particular, is of functional relevance for immune modulation in this assay setup, highlighting the need to study IgA mediated immune response.
Collapse
Affiliation(s)
- Fabian Bohländer
- Department of Analytical Development and Validation, Corporate R&D, Biotest AG, Dreieich, Germany
- Corporate R&D, Biotest AG, Dreieich, Germany
| | - Dennis Riehl
- Department of Analytical Development and Validation, Corporate R&D, Biotest AG, Dreieich, Germany
- Corporate R&D, Biotest AG, Dreieich, Germany
| | - Sabrina Weißmüller
- Corporate R&D, Biotest AG, Dreieich, Germany
- Department of Translational Research, Preclinical Research, Corporate R&D, Biotest AG, Dreieich, Germany
| | - Marcus Gutscher
- Department of Analytical Development and Validation, Corporate R&D, Biotest AG, Dreieich, Germany
- Corporate R&D, Biotest AG, Dreieich, Germany
| | | | - Stefanie Faust
- Department of Analytical Development and Validation, Corporate R&D, Biotest AG, Dreieich, Germany
- Corporate R&D, Biotest AG, Dreieich, Germany
| |
Collapse
|
28
|
Immohr MB, Akhyari P, Aubin H, Westenfeld R, Mehdiani A, Bruno RR, Sipahi NF, Erbel-Khurtsidze S, Reinecke P, Tudorache I, Lichtenberg A, Boeken U. Treatment of donor-specific antibody-mediated rejection after heart transplantation by IgM-enriched human immunoglobulin. ESC Heart Fail 2021; 8:3413-3417. [PMID: 33969938 PMCID: PMC8318410 DOI: 10.1002/ehf2.13409] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/07/2021] [Accepted: 04/24/2021] [Indexed: 12/25/2022] Open
Abstract
Antibody‐mediated graft rejection caused by donor‐specific antibodies (DSA‐MR) remains a serious problem after heart transplantation (HTx). IgM‐enriched human intravenous immunoglobulin (IGM‐IVIG) consists of 76% IgG, 12% IgM, and 12% IgA and provides a new multifactorial approach for DSA‐MR. Between 2017 and 2020, four (P1–4) of 102 patients developed DSA‐MR after HTx in our department and were repetitively treated with IGM‐IVIG in combination with anti‐thymocyte globulin. While in P1 and P4, DSA‐MR occurred within the early post‐operative interval, P2 and P3 developed DSA‐MR approximately 1 year after transplantation. An impairment of ventricular function was observed in three of four patients. Furthermore, P1 and P4 suffered from malign ventricular arrhythmias. After the application of IGM‐IVIG, the ventricular function recovered, and all patients could be discharged from the hospital. As part of a multifactorial therapeutic approach, treatment with IGM‐IVIG seems to be a safe and effective strategy to address DSA‐MR.
Collapse
Affiliation(s)
- Moritz Benjamin Immohr
- Department of Cardiac Surgery, Medical Faculty, Heinrich-Heine-University Medical School, Moorenstraße 5, Duesseldorf, 40225, Germany
| | - Payam Akhyari
- Department of Cardiac Surgery, Medical Faculty, Heinrich-Heine-University Medical School, Moorenstraße 5, Duesseldorf, 40225, Germany
| | - Hug Aubin
- Department of Cardiac Surgery, Medical Faculty, Heinrich-Heine-University Medical School, Moorenstraße 5, Duesseldorf, 40225, Germany
| | - Ralf Westenfeld
- Department of Cardiology, Pulmonology and Vascular Medicine, Heinrich-Heine-University Medical School, Duesseldorf, Germany
| | - Arash Mehdiani
- Department of Cardiac Surgery, Medical Faculty, Heinrich-Heine-University Medical School, Moorenstraße 5, Duesseldorf, 40225, Germany
| | - Raphael Romano Bruno
- Department of Cardiology, Pulmonology and Vascular Medicine, Heinrich-Heine-University Medical School, Duesseldorf, Germany
| | - Nihat Firat Sipahi
- Department of Cardiac Surgery, Medical Faculty, Heinrich-Heine-University Medical School, Moorenstraße 5, Duesseldorf, 40225, Germany
| | - Sophiko Erbel-Khurtsidze
- Department of Cardiac Surgery, Medical Faculty, Heinrich-Heine-University Medical School, Moorenstraße 5, Duesseldorf, 40225, Germany
| | - Petra Reinecke
- Institute of Pathology, Heinrich-Heine-University Medical School, Duesseldorf, Germany
| | - Igor Tudorache
- Department of Cardiac Surgery, Medical Faculty, Heinrich-Heine-University Medical School, Moorenstraße 5, Duesseldorf, 40225, Germany
| | - Artur Lichtenberg
- Department of Cardiac Surgery, Medical Faculty, Heinrich-Heine-University Medical School, Moorenstraße 5, Duesseldorf, 40225, Germany
| | - Udo Boeken
- Department of Cardiac Surgery, Medical Faculty, Heinrich-Heine-University Medical School, Moorenstraße 5, Duesseldorf, 40225, Germany
| |
Collapse
|
29
|
Estruch J, Johnson J, Ford S, Yoshimoto S, Mills T, Bergman P. Utility of the combined use of 3 serologic markers in the diagnosis and monitoring of chronic enteropathies in dogs. J Vet Intern Med 2021; 35:1306-1315. [PMID: 33960552 PMCID: PMC8163139 DOI: 10.1111/jvim.16132] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 03/26/2021] [Accepted: 04/07/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Dogs with chronic enteropathies (CE) displayed elevated IgA seropositivity against specific markers that can be used to develop a novel test. OBJECTIVE To assess a multivariate test to aid diagnosis of CE in dogs and to monitor treatment-related responses. ANIMALS One hundred fifty-seven dogs with CE/inflammatory bowel disease (IBD), 24 dogs non-IBD gastrointestinal disorders, and 33 normal dogs. METHODS Prospective, multicenter, clinical study that enrolled dogs with gastrointestinal disorders. Serum sample collected at enrollment and up to 3 months follow-up measuring OmpC (ACA), canine calprotectin (ACNA), and gliadin-derived peptides (AGA) by ELISA. RESULTS Seropositivity was higher in CE/IBD than normal dogs (66% vs 9% for ACA; 55% vs 15% for ACNA; and 75% vs 6% for AGA; P < .001). When comparing CE/IBD with non-IBD disease, ACA and ACNA displayed discriminating properties (66%, 55% vs 12.5%, 29% respectively) while AGA separated CE from normal cohorts (54% vs 6%). A 3-marker algorithm at cutoff of ACA > 15, ACNA > 6, AGA > 60 differentiates CE/IBD and normal dogs with 90% sensitivity and 96% specificity; and CE/IBD and non-IBD dogs with 80% sensitivity and 86% specificity. Titers decreased after treatment (47%-99% in ACA, 13%-88% in ACNA, and 30%-85% in AGA), changes that were concurrent with clinical improvements. CONCLUSION AND CLINICAL IMPORTANCE An assay based on combined measurements of ACA, ACNA, and AGA is useful as a noninvasive diagnostic test to distinguish dogs with CE/IBD. The test also has the potential to monitor response to treatment.
Collapse
Affiliation(s)
| | | | - Sarah Ford
- Department of Internal MedicineBlue Pearl SpecialtyScottsdaleArizonaUSA
| | - Sean Yoshimoto
- Department of Internal MedicineWest LA Animal HospitalLos AngelesCaliforniaUSA
| | - Tracy Mills
- Department of Clinical StudiesWest LA Animal HospitalLos AngelesCaliforniaUSA
| | - Philip Bergman
- Department of Internal MedicineKatonah Bedford Veterinary CenterNew YorkNew YorkUSA
| |
Collapse
|
30
|
Chin-Fatt A, Saberianfar R, Menassa R. A Rationally Designed Bovine IgA Fc Scaffold Enhances in planta Accumulation of a V HH-Fc Fusion Without Compromising Binding to Enterohemorrhagic E. coli. FRONTIERS IN PLANT SCIENCE 2021; 12:651262. [PMID: 33936135 PMCID: PMC8079772 DOI: 10.3389/fpls.2021.651262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 03/22/2021] [Indexed: 06/12/2023]
Abstract
We previously isolated a single domain antibody (VHH) that binds Enterohemorrhagic Escherichia coli (EHEC) with the end-goal being the enteromucosal passive immunization of cattle herds. To improve the yield of a chimeric fusion of the VHH with an IgA Fc, we employed two rational design strategies, supercharging and introducing de novo disulfide bonds, on the bovine IgA Fc component of the chimera. After mutagenizing the Fc, we screened for accumulation levels after transient transformation in Nicotiana benthamiana leaves. We identified and characterized five supercharging and one disulfide mutant, termed '(5 + 1)Fc', that improve accumulation in comparison to the native Fc. Combining all these mutations is associated with a 32-fold increase of accumulation for the Fc alone, from 23.9 mg/kg fresh weight (FW) to 599.5 mg/kg FW, as well as a twenty-fold increase when fused to a VHH that binds EHEC, from 12.5 mg/kg FW tissue to 236.2 mg/kg FW. Co-expression of native or mutated VHH-Fc with bovine joining chain (JC) and bovine secretory component (SC) followed by co-immunoprecipitation suggests that the stabilizing mutations do not interfere with the capacity of VHH-Fc to assemble with JC and FC into a secretory IgA. Both the native and the mutated VHH-Fc similarly neutralized the ability of four of the seven most prevalent EHEC strains (O157:H7, O26:H11, O111:Hnm, O145:Hnm, O45:H2, O121:H19 and O103:H2), to adhere to HEp-2 cells as visualized by immunofluorescence microscopy and quantified by fluorometry. These results collectively suggest that supercharging and disulfide bond tethering on a Fc chain can effectively improve accumulation of a VHH-Fc fusion without impacting VHH functionality.
Collapse
Affiliation(s)
- Adam Chin-Fatt
- Agriculture and Agri-Food Canada, London Research and Development Centre, London, ON, Canada
- Department of Biology, University of Western Ontario, London, ON, Canada
| | - Reza Saberianfar
- Agriculture and Agri-Food Canada, London Research and Development Centre, London, ON, Canada
| | - Rima Menassa
- Agriculture and Agri-Food Canada, London Research and Development Centre, London, ON, Canada
- Department of Biology, University of Western Ontario, London, ON, Canada
| |
Collapse
|
31
|
Bhaskara V, Leal MT, Seigner J, Friedrich T, Kreidl E, Gadermaier E, Tesarz M, Rogalli A, Stangl L, Wallwitz J, Hammel K, Rothbauer M, Moll H, Ertl P, Hahn R, Himmler G, Bauer A, Casanova E. Efficient production of recombinant secretory IgA against Clostridium difficile toxins in CHO-K1 cells. J Biotechnol 2021; 331:1-13. [PMID: 33689865 DOI: 10.1016/j.jbiotec.2021.02.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 01/25/2021] [Accepted: 02/22/2021] [Indexed: 12/29/2022]
Abstract
Despite the essential role secretory IgAs play in the defense against pathogenic invasion and the proposed value of recombinant secretory IgAs as novel therapeutics, currently there are no IgA-based therapies in clinics. Secretory IgAs are complex molecules and the major bottleneck limiting their therapeutic potential is a reliable recombinant production system. In this report, we addressed this issue and established a fast and robust production method for secretory IgAs in CHO-K1 cells using BAC-based expression vectors. As a proof of principle, we produced IgAs against Clostridium difficile toxins TcdA and TcdB. Recombinant secretory IgAs produced using our expression system showed comparable titers to IgGs, widely used as therapeutic biologicals. Importantly, secretory IgAs produced using our method were functional and could efficiently neutralize Clostridium difficile toxins TcdA and TcdB. These results show that recombinant secretory IgAs can be efficiently produced, thus opening the possibility to use them as therapeutic agents in clinics.
Collapse
Affiliation(s)
- Venugopal Bhaskara
- Department of Physiology, Center of Physiology and Pharmacology and Comprehensive Cancer Center (CCC), Medical University of Vienna, 1090 Vienna, Austria.
| | - Maria Trinidad Leal
- Department of Physiology, Center of Physiology and Pharmacology and Comprehensive Cancer Center (CCC), Medical University of Vienna, 1090 Vienna, Austria
| | - Jacqueline Seigner
- Department of Physiology, Center of Physiology and Pharmacology and Comprehensive Cancer Center (CCC), Medical University of Vienna, 1090 Vienna, Austria
| | - Theresa Friedrich
- Department of Physiology, Center of Physiology and Pharmacology and Comprehensive Cancer Center (CCC), Medical University of Vienna, 1090 Vienna, Austria
| | | | | | | | | | - Laura Stangl
- Department of Physiology, Center of Physiology and Pharmacology and Comprehensive Cancer Center (CCC), Medical University of Vienna, 1090 Vienna, Austria
| | | | - Katharina Hammel
- Department for Biotechnology, University of Natural Resources and Life Sciences Vienna, 1190 Vienna, Austria
| | - Mario Rothbauer
- Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Faculty of Technical Chemistry, 1060 Vienna, Austria
| | - Herwig Moll
- Department of Physiology, Center of Physiology and Pharmacology and Comprehensive Cancer Center (CCC), Medical University of Vienna, 1090 Vienna, Austria
| | - Peter Ertl
- Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Faculty of Technical Chemistry, 1060 Vienna, Austria
| | - Rainer Hahn
- Department for Biotechnology, University of Natural Resources and Life Sciences Vienna, 1190 Vienna, Austria
| | | | - Anton Bauer
- Department of Physiology, Center of Physiology and Pharmacology and Comprehensive Cancer Center (CCC), Medical University of Vienna, 1090 Vienna, Austria; The Antibody Lab GmbH, 1210 Vienna, Austria.
| | - Emilio Casanova
- Department of Physiology, Center of Physiology and Pharmacology and Comprehensive Cancer Center (CCC), Medical University of Vienna, 1090 Vienna, Austria.
| |
Collapse
|
32
|
Maruthachalam BV, Zwolak A, Lin-Schmidt X, Keough E, Tamot N, Venkataramani S, Geist B, Singh S, Ganesan R. Discovery and characterization of single-domain antibodies for polymeric Ig receptor-mediated mucosal delivery of biologics. MAbs 2021; 12:1708030. [PMID: 31906797 PMCID: PMC6973331 DOI: 10.1080/19420862.2019.1708030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mucosal immunity is dominated by secretory IgA and IgM, although these are less favorable compared to IgG molecules for therapeutic development. Polymeric IgA and IgM are actively transported across the epithelial barrier via engagement of the polymeric Ig receptor (pIgR), but IgG molecules lack a lumen-targeted active transport mechanism, resulting in poor biodistribution of IgG therapeutics in mucosal tissues. In this work, we describe the discovery and characterization of single-domain antibodies (VHH) that engage pIgR and undergo transepithelial transport across the mucosal epithelium. The anti-pIgR VHH panel displayed a broad range of biophysical characteristics, epitope diversity, IgA competition profiles and transcytosis activity in cell and human primary lung tissue models. Making use of this diverse VHH panel, we studied the relationship between biophysical and functional properties of anti-pIgR binders targeting different domains and epitopes of pIgR. These VHH molecules will serve as excellent tools for studying pIgR-mediated transport of biologics and for delivering multispecific IgG antibodies into mucosal lumen, where they can target and neutralize mucosal antigens.
Collapse
Affiliation(s)
| | - Adam Zwolak
- Janssen BioTherapeutics, Janssen Research and Development, Spring House, PA, USA
| | - Xiefan Lin-Schmidt
- Janssen BioTherapeutics, Janssen Research and Development, Spring House, PA, USA
| | - Edward Keough
- Janssen BioTherapeutics, Janssen Research and Development, Spring House, PA, USA
| | - Ninkka Tamot
- Janssen BioTherapeutics, Janssen Research and Development, Spring House, PA, USA
| | - Sathya Venkataramani
- Janssen BioTherapeutics, Janssen Research and Development, Spring House, PA, USA
| | - Brian Geist
- Janssen BioTherapeutics, Janssen Research and Development, Spring House, PA, USA
| | - Sanjaya Singh
- Janssen BioTherapeutics, Janssen Research and Development, Spring House, PA, USA
| | - Rajkumar Ganesan
- Janssen BioTherapeutics, Janssen Research and Development, Spring House, PA, USA
| |
Collapse
|
33
|
van Gool MMJ, van Egmond M. IgA and FcαRI: Versatile Players in Homeostasis, Infection, and Autoimmunity. Immunotargets Ther 2021; 9:351-372. [PMID: 33447585 PMCID: PMC7801909 DOI: 10.2147/itt.s266242] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/17/2020] [Indexed: 12/11/2022] Open
Abstract
Mucosal surfaces constitute the frontiers of the body and are the biggest barriers of our body for the outside world. Immunoglobulin A (IgA) is the most abundant antibody class present at these sites. It passively contributes to mucosal homeostasis via immune exclusion maintaining a tight balance between tolerating commensals and providing protection against pathogens. Once pathogens have succeeded in invading the epithelial barriers, IgA has an active role in host-pathogen defense by activating myeloid cells through divers receptors, including its Fc receptor, FcαRI (CD89). To evade elimination, several pathogens secrete proteins that interfere with either IgA neutralization or FcαRI-mediated immune responses, emphasizing the importance of IgA-FcαRI interactions in preventing infection. Depending on the IgA form, either anti- or pro-inflammatory responses can be induced. Moreover, the presence of excessive IgA immune complexes can result in continuous FcαRI-mediated activation of myeloid cells, potentially leading to severe tissue damage. On the one hand, enhancing pathogen-specific mucosal and systemic IgA by vaccination may increase protective immunity against infectious diseases. On the other hand, interfering with the IgA-FcαRI axis by monovalent targeting or blocking FcαRI may resolve IgA-induced inflammation and tissue damage. This review describes the multifaceted role of FcαRI as immune regulator between anti- and pro-inflammatory responses of IgA, and addresses potential novel therapeutic strategies that target FcαRI in disease. ![]()
Point your SmartPhone at the code above. If you have a QR code reader the video abstract will appear. Or use: https://youtu.be/xlijXy5W0xA
Collapse
Affiliation(s)
- Melissa Maria Johanna van Gool
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, Netherlands.,Amsterdam institute for Infection and Immunity, Amsterdam UMC, Amsterdam, Netherlands
| | - Marjolein van Egmond
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, Netherlands.,Amsterdam institute for Infection and Immunity, Amsterdam UMC, Amsterdam, Netherlands.,Department of Surgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
34
|
Linking molecular targets of Cd in the bloodstream to organ-based adverse health effects. J Inorg Biochem 2020; 216:111279. [PMID: 33413916 DOI: 10.1016/j.jinorgbio.2020.111279] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/05/2020] [Accepted: 10/05/2020] [Indexed: 12/25/2022]
Abstract
The chronic exposure of human populations to toxic metals remains a global public health concern. Although chronic Cd exposure is linked to kidney damage, osteoporosis and cancer, the underlying biomolecular mechanisms remain incompletely understood. Since other diseases could also be causally linked to chronic Cd exposure, a systems toxicology-based approach is needed to gain new insight into the underlying exposure-disease relationship. This approach requires one to integrate the cascade of dynamic bioinorganic chemistry events that unfold in the bloodstream after Cd enters with toxicological events that unfold in target organs over time. To this end, we have conducted a systematic literature search to identify all molecular targets of Cd in plasma and in red blood cells (RBCs). Based on this information it is impossible to describe the metabolism of Cd and the toxicological relevance of it binding to molecular targets in/on RBCs is elusive. Perhaps most importantly, the role that peptides, amino acids and inorganic ions, including HCO3-, Cl- and HSeO3- play in terms of mediating the translocation of Cd to target organs and its detoxification is poorly understood. Causally linking human exposure to this metal with diseases requires a much better integration of the bioinorganic chemistry of Cd that unfolds in the bloodstream with target organs. This from a public health point of view important goal will require collaborations between scientists from different disciplines to untangle the complex mechanisms which causally link Cd exposure to disease.
Collapse
|
35
|
Ejemel M, Li Q, Hou S, Schiller ZA, Tree JA, Wallace A, Amcheslavsky A, Kurt Yilmaz N, Buttigieg KR, Elmore MJ, Godwin K, Coombes N, Toomey JR, Schneider R, Ramchetty AS, Close BJ, Chen DY, Conway HL, Saeed M, Ganesa C, Carroll MW, Cavacini LA, Klempner MS, Schiffer CA, Wang Y. A cross-reactive human IgA monoclonal antibody blocks SARS-CoV-2 spike-ACE2 interaction. Nat Commun 2020; 11:4198. [PMID: 32826914 PMCID: PMC7442812 DOI: 10.1038/s41467-020-18058-8] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 08/03/2020] [Indexed: 01/19/2023] Open
Abstract
COVID-19 caused by SARS-CoV-2 has become a global pandemic requiring the development of interventions for the prevention or treatment to curtail mortality and morbidity. No vaccine to boost mucosal immunity, or as a therapeutic, has yet been developed to SARS-CoV-2. In this study, we discover and characterize a cross-reactive human IgA monoclonal antibody, MAb362. MAb362 binds to both SARS-CoV and SARS-CoV-2 spike proteins and competitively blocks ACE2 receptor binding, by overlapping the ACE2 structural binding epitope. Furthermore, MAb362 IgA neutralizes both pseudotyped SARS-CoV and SARS-CoV-2 in 293 cells expressing ACE2. When converted to secretory IgA, MAb326 also neutralizes authentic SARS-CoV-2 virus while the IgG isotype shows no neutralization. Our results suggest that SARS-CoV-2 specific IgA antibodies, such as MAb362, may provide effective immunity against SARS-CoV-2 by inducing mucosal immunity within the respiratory system, a potentially critical feature of an effective vaccine. Here, Ejemel et al. report the identification and characterization of a cross-neutralizing human IgA monoclonal antibody, named MAb362, that binds the receptor-binding domain of SARS-CoV-2 Spike, blocking its interaction with the ACE2 host receptor.
Collapse
Affiliation(s)
- Monir Ejemel
- MassBiologics of the University of Massachusetts Medical School, Boston, MA, USA
| | - Qi Li
- MassBiologics of the University of Massachusetts Medical School, Boston, MA, USA
| | - Shurong Hou
- Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Boston, MA, USA
| | - Zachary A Schiller
- MassBiologics of the University of Massachusetts Medical School, Boston, MA, USA
| | - Julia A Tree
- National Infection Service, Public Health England, Porton Down, Salisbury, Wiltshire, UK
| | - Aaron Wallace
- MassBiologics of the University of Massachusetts Medical School, Boston, MA, USA
| | - Alla Amcheslavsky
- MassBiologics of the University of Massachusetts Medical School, Boston, MA, USA
| | - Nese Kurt Yilmaz
- Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Boston, MA, USA
| | - Karen R Buttigieg
- National Infection Service, Public Health England, Porton Down, Salisbury, Wiltshire, UK
| | - Michael J Elmore
- National Infection Service, Public Health England, Porton Down, Salisbury, Wiltshire, UK
| | - Kerry Godwin
- National Infection Service, Public Health England, Porton Down, Salisbury, Wiltshire, UK
| | - Naomi Coombes
- National Infection Service, Public Health England, Porton Down, Salisbury, Wiltshire, UK
| | - Jacqueline R Toomey
- MassBiologics of the University of Massachusetts Medical School, Boston, MA, USA
| | - Ryan Schneider
- MassBiologics of the University of Massachusetts Medical School, Boston, MA, USA
| | - Anudeep S Ramchetty
- MassBiologics of the University of Massachusetts Medical School, Boston, MA, USA
| | - Brianna J Close
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Da-Yuan Chen
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Hasahn L Conway
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Mohsan Saeed
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Chandrashekar Ganesa
- MassBiologics of the University of Massachusetts Medical School, Boston, MA, USA
| | - Miles W Carroll
- National Infection Service, Public Health England, Porton Down, Salisbury, Wiltshire, UK
| | - Lisa A Cavacini
- MassBiologics of the University of Massachusetts Medical School, Boston, MA, USA.
| | - Mark S Klempner
- MassBiologics of the University of Massachusetts Medical School, Boston, MA, USA.
| | - Celia A Schiffer
- Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Boston, MA, USA.
| | - Yang Wang
- MassBiologics of the University of Massachusetts Medical School, Boston, MA, USA.
| |
Collapse
|
36
|
Ejemel M, Li Q, Hou S, Schiller ZA, Wallace AL, Amcheslavsky A, Yilmaz NK, Toomey JR, Schneider R, Close BJ, Chen DY, Conway HL, Mohsan S, Cavacini LA, Klempner MS, Schiffer CA, Wang Y. IgA MAb blocks SARS-CoV-2 Spike-ACE2 interaction providing mucosal immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020. [PMID: 32511396 DOI: 10.1101/2020.05.15.096719] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
COVID-19 caused by SARS-CoV-2 has become a global pandemic requiring the development of interventions for the prevention or treatment to curtail mortality and morbidity. No vaccine to boost mucosal immunity or as a therapeutic has yet been developed to SARS-CoV-2. In this study we discover and characterize a cross-reactive human IgA monoclonal antibody, MAb362. MAb362 binds to both SARS-CoV and SARS-CoV-2 spike proteins and competitively blocks hACE2 receptor binding, by completely overlapping the hACE2 structural binding epitope. Furthermore, MAb362 IgA neutralizes both pseudotyped SARS-CoV and SARS-CoV-2 in human epithelial cells expressing hACE2. SARS-CoV-2 specific IgA antibodies, such as MAb362, may provide effective immunity against SARS-CoV-2 by inducing mucosal immunity within the respiratory system, a potentially critical feature of an effective vaccine.
Collapse
|
37
|
Langel SN, Otero CE, Martinez DR, Permar SR. Maternal gatekeepers: How maternal antibody Fc characteristics influence passive transfer and infant protection. PLoS Pathog 2020; 16:e1008303. [PMID: 32214394 PMCID: PMC7098569 DOI: 10.1371/journal.ppat.1008303] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Stephanie N. Langel
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, United States of America
- * E-mail: (SNL); (SRR)
| | - Claire E. Otero
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, United States of America
| | - David R. Martinez
- Department of Epidemiology, University of North Carolina at Chapel Hill School of Public Health, Chapel Hill, North Carolina, United States of America
| | - Sallie R. Permar
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, United States of America
- * E-mail: (SNL); (SRR)
| |
Collapse
|
38
|
Davis SK, Selva KJ, Kent SJ, Chung AW. Serum IgA Fc effector functions in infectious disease and cancer. Immunol Cell Biol 2020; 98:276-286. [PMID: 31785006 PMCID: PMC7217208 DOI: 10.1111/imcb.12306] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 11/24/2019] [Accepted: 11/26/2019] [Indexed: 12/18/2022]
Abstract
Immunoglobulin (Ig) A is the most abundant antibody isotype present at mucosal surfaces and the second most abundant in human serum. In addition to preventing pathogen entry at mucosal surfaces, IgA can control and eradicate bacterial and viral infections through a variety of antibody‐mediated innate effector cell mechanisms. The role of mucosal IgA in infection (e.g. neutralization) and in inflammatory homeostasis (e.g. allergy and autoimmunity) has been extensively investigated; by contrast, serum IgA is comparatively understudied. IgA binding to fragment crystallizable alpha receptor plays a dual role in the activation and inhibition of innate effector cell functions. Mounting evidence suggests that serum IgA induces potent effector functions against various bacterial and some viral infections including Neisseria meningitidis and rotavirus. Furthermore, in the era of immunotherapy, serum IgA provides an interesting alternative to classical IgG monoclonal antibodies to treat cancer and infectious pathogens. Here we discuss the role of serum IgA in infectious diseases with reference to bacterial and viral infections and the potential for IgA as a monoclonal antibody therapy.
Collapse
Affiliation(s)
- Samantha K Davis
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, Australia
| | - Kevin J Selva
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, Australia.,Melbourne Sexual Health Centre, Infectious Diseases Department, Alfred Health, Central Clinical School, Monash University, Melbourne, VIC, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, VIC, Australia
| | - Amy W Chung
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
39
|
de Sousa-Pereira P, Woof JM. IgA: Structure, Function, and Developability. Antibodies (Basel) 2019; 8:antib8040057. [PMID: 31817406 PMCID: PMC6963396 DOI: 10.3390/antib8040057] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 11/24/2019] [Accepted: 11/28/2019] [Indexed: 02/07/2023] Open
Abstract
Immunoglobulin A (IgA) plays a key role in defending mucosal surfaces against attack by infectious microorganisms. Such sites present a major site of susceptibility due to their vast surface area and their constant exposure to ingested and inhaled material. The importance of IgA to effective immune defence is signalled by the fact that more IgA is produced than all the other immunoglobulin classes combined. Indeed, IgA is not just the most prevalent antibody class at mucosal sites, but is also present at significant concentrations in serum. The unique structural features of the IgA heavy chain allow IgA to polymerise, resulting in mainly dimeric forms, along with some higher polymers, in secretions. Both serum IgA, which is principally monomeric, and secretory forms of IgA are capable of neutralising and removing pathogens through a range of mechanisms, including triggering the IgA Fc receptor known as FcαRI or CD89 on phagocytes. The effectiveness of these elimination processes is highlighted by the fact that various pathogens have evolved mechanisms to thwart such IgA-mediated clearance. As the structure–function relationships governing the varied capabilities of this immunoglobulin class come into increasingly clear focus, and means to circumvent any inherent limitations are developed, IgA-based monoclonal antibodies are set to emerge as new and potent options in the therapeutic arena.
Collapse
Affiliation(s)
- Patrícia de Sousa-Pereira
- School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
- CIBIO-InBIO, Campus Agrário de Vairão, University of Porto, 4485-661 Vairão, Portugal
| | - Jenny M. Woof
- School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
- Correspondence: ; Tel.: +44-1382-383389
| |
Collapse
|
40
|
Lombana TN, Rajan S, Zorn JA, Mandikian D, Chen EC, Estevez A, Yip V, Bravo DD, Phung W, Farahi F, Viajar S, Lee S, Gill A, Sandoval W, Wang J, Ciferri C, Boswell CA, Matsumoto ML, Spiess C. Production, characterization, and in vivo half-life extension of polymeric IgA molecules in mice. MAbs 2019; 11:1122-1138. [PMID: 31122132 PMCID: PMC6748581 DOI: 10.1080/19420862.2019.1622940] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
IgA antibodies have broad potential as a novel therapeutic platform based on their superior receptor-mediated cytotoxic activity, potent neutralization of pathogens, and ability to transcytose across mucosal barriers via polymeric immunoglobulin receptor (pIgR)-mediated transport, compared to traditional IgG-based drugs. However, the transition of IgA into clinical development has been challenged by complex expression and characterization, as well as rapid serum clearance that is thought to be mediated by glycan receptor scavenging of recombinantly produced IgA monomer bearing incompletely sialylated N-linked glycans. Here, we present a comprehensive biochemical, biophysical, and structural characterization of recombinantly produced monomeric, dimeric and polymeric human IgA. We further explore two strategies to overcome the rapid serum clearance of polymeric IgA: removal of all N-linked glycosylation sites creating an aglycosylated polymeric IgA and engineering in FcRn binding with the generation of a polymeric IgG-IgA Fc fusion. While previous reports and the results presented in this study indicate that glycan-mediated clearance plays a major role for monomeric IgA, systemic clearance of polymeric IgA in mice is predominantly controlled by mechanisms other than glycan receptor clearance, such as pIgR-mediated transcytosis. The developed IgA platform now provides the potential to specifically target pIgR expressing tissues, while maintaining low systemic exposure.
Collapse
Affiliation(s)
- T Noelle Lombana
- a Department of Antibody Engineering, Genentech Inc., South San Francisco , CA , USA
| | - Sharmila Rajan
- b Department of Preclinical and Translational Pharmacokinetics, Genentech Inc., South San Francisco , CA , USA
| | - Julie A Zorn
- c Department of Structural Biology, Genentech Inc., South San Francisco , CA , USA
| | - Danielle Mandikian
- b Department of Preclinical and Translational Pharmacokinetics, Genentech Inc., South San Francisco , CA , USA
| | - Eugene C Chen
- d Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco , CA , USA
| | - Alberto Estevez
- c Department of Structural Biology, Genentech Inc., South San Francisco , CA , USA
| | - Victor Yip
- b Department of Preclinical and Translational Pharmacokinetics, Genentech Inc., South San Francisco , CA , USA
| | - Daniel D Bravo
- e Department of Biochemical and Cellular Pharmacology, Genentech Inc., South San Francisco , CA , USA
| | - Wilson Phung
- f Department of Microchemistry, Proteomics and Lipidomics, Genentech Inc. , South San Francisco , CA , USA
| | - Farzam Farahi
- a Department of Antibody Engineering, Genentech Inc., South San Francisco , CA , USA
| | - Sharon Viajar
- a Department of Antibody Engineering, Genentech Inc., South San Francisco , CA , USA
| | - Sophia Lee
- a Department of Antibody Engineering, Genentech Inc., South San Francisco , CA , USA
| | - Avinash Gill
- a Department of Antibody Engineering, Genentech Inc., South San Francisco , CA , USA
| | - Wendy Sandoval
- f Department of Microchemistry, Proteomics and Lipidomics, Genentech Inc. , South San Francisco , CA , USA
| | - Jianyong Wang
- e Department of Biochemical and Cellular Pharmacology, Genentech Inc., South San Francisco , CA , USA
| | - Claudio Ciferri
- c Department of Structural Biology, Genentech Inc., South San Francisco , CA , USA
| | - C Andrew Boswell
- b Department of Preclinical and Translational Pharmacokinetics, Genentech Inc., South San Francisco , CA , USA
| | - Marissa L Matsumoto
- c Department of Structural Biology, Genentech Inc., South San Francisco , CA , USA
| | - Christoph Spiess
- a Department of Antibody Engineering, Genentech Inc., South San Francisco , CA , USA
| |
Collapse
|
41
|
|
42
|
Breedveld A, van Egmond M. IgA and FcαRI: Pathological Roles and Therapeutic Opportunities. Front Immunol 2019; 10:553. [PMID: 30984170 PMCID: PMC6448004 DOI: 10.3389/fimmu.2019.00553] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 03/01/2019] [Indexed: 12/12/2022] Open
Abstract
Immunoglobulin A (IgA) is the most abundant antibody class present at mucosal surfaces. The production of IgA exceeds the production of all other antibodies combined, supporting its prominent role in host-pathogen defense. IgA closely interacts with the intestinal microbiota to enhance its diversity, and IgA has a passive protective role via immune exclusion. Additionally, inhibitory ITAMi signaling via the IgA Fc receptor (FcαRI; CD89) by monomeric IgA may play a role in maintaining homeostatic conditions. By contrast, IgA immune complexes (e.g., opsonized pathogens) potently activate immune cells via cross-linking FcαRI, thereby inducing pro-inflammatory responses resulting in elimination of pathogens. The importance of IgA in removal of pathogens is emphasized by the fact that several pathogens developed mechanisms to break down IgA or evade FcαRI-mediated activation of immune cells. Augmented or aberrant presence of IgA immune complexes can result in excessive neutrophil activation, potentially leading to severe tissue damage in multiple inflammatory, or autoimmune diseases. Influencing IgA or FcαRI-mediated functions therefore provides several therapeutic possibilities. On the one hand (passive) IgA vaccination strategies can be developed for protection against infections. Furthermore, IgA monoclonal antibodies that are directed against tumor antigens may be effective as cancer treatment. On the other hand, induction of ITAMi signaling via FcαRI may reduce allergy or inflammation, whereas blocking FcαRI with monoclonal antibodies, or peptides may resolve IgA-induced tissue damage. In this review both (patho)physiological roles as well as therapeutic possibilities of the IgA-FcαRI axis are addressed.
Collapse
Affiliation(s)
- Annelot Breedveld
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Amsterdam, Netherlands
- Amsterdam Infection and Immunity Institute, Amsterdam UMC, Amsterdam, Netherlands
| | - Marjolein van Egmond
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Amsterdam, Netherlands
- Amsterdam Infection and Immunity Institute, Amsterdam UMC, Amsterdam, Netherlands
- Department of Surgery, Amsterdam UMC, Amsterdam, Netherlands
| |
Collapse
|
43
|
Frontera ED, Khansa RM, Schalk DL, Leakan LE, Guerin-Edbauer TJ, Ratnam M, Gorski DH, Speyer CL. IgA Fc-folate conjugate activates and recruits neutrophils to directly target triple-negative breast cancer cells. Breast Cancer Res Treat 2018; 172:551-560. [PMID: 30155754 DOI: 10.1007/s10549-018-4941-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 08/25/2018] [Indexed: 10/28/2022]
Abstract
PURPOSE According to the American Cancer Society, 1 in 8 women in the U.S. will develop breast cancer, with triple-negative breast cancer (TNBC) comprising 15-20% of all breast cancer cases. TNBC is an aggressive subtype due to its high metastatic potential and lack of targeted therapy. Recently, folate receptor alpha (FRA) is found to be expressed on 80% of TNBC with high expression correlating with poor prognosis. In this study, we examined whether binding IgA Fc-folate molecules to FRA receptors on TNBC cells can elicit and induce neutrophils (PMNs), by binding their FcαR1 receptors, to destroy TNBC cells. METHODS FRA was analyzed on TNBC cells and binding assays were performed using 3H-folate. Fc-folate was synthesized by linking Fc fragments of IgA via amine groups to folate. Binding specificity and antibody-dependent cellular cytotoxicity (ADCC) potential of Fc-folate to FcαR1 were confirmed by measuring PMN adhesion and myeloperoxidase (MPO) release in a cell-based ELISA. Fc-folate binding to FRA-expressing TNBC cells inducing PMNs to destroy these cells was determined using 51Cr-release and calcein-labeling assays. RESULTS Our results demonstrate expression of FRA on TNBC cells at levels consistent with folate binding. Fc-folate binds with high affinity to FRA compared to whole IgA-folate and induces MPO release from PMN when bound to FcαR1. Fc-folate inhibited binding of 3H-folate to TNBC cells and induced significant cell lysis of TNBC cells when incubated in the presence of PMNs. CONCLUSION These findings support the hypothesis that an IgA Fc-folate conjugate can destroy TNBC cells by eliciting PMN-mediated ADCC.
Collapse
Affiliation(s)
- Eric D Frontera
- College of Osteopathic Medicine, Michigan State University, 4707 St. Antoine, Box 402, Detroit, MI, 48201, USA
| | - Rafa M Khansa
- College of Osteopathic Medicine, Michigan State University, 4707 St. Antoine, Box 402, Detroit, MI, 48201, USA
| | - Dana L Schalk
- Department of Hematology/Oncology, University of Virginia Cancer Center, 1300 Jefferson Park Ave., I MSB 7191 West Complex I, Charlottesville, VA, 22903, USA
| | - Lauren E Leakan
- Wayne State University, 42 West Warren Avenue, Detroit, MI, 48202, USA
| | - Tracey J Guerin-Edbauer
- Michael and Marian Ilitch Department of Surgery, Wayne State University School of Medicine, 4100 John R St., Mailcode HW08AO, Detroit, MI, 48201, USA
| | - Manohar Ratnam
- Department of Oncology, Wayne State University School of Medicine, 4100 John R St., Detroit, MI, 48201, USA.,Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, 4100 John R St., Detroit, MI, 48201, USA
| | - David H Gorski
- Michael and Marian Ilitch Department of Surgery, Wayne State University School of Medicine, 4100 John R St., Mailcode HW08AO, Detroit, MI, 48201, USA.,Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, 4100 John R St., Detroit, MI, 48201, USA
| | - Cecilia L Speyer
- Michael and Marian Ilitch Department of Surgery, Wayne State University School of Medicine, 4100 John R St., Mailcode HW08AO, Detroit, MI, 48201, USA. .,Tumor Microenvironment Program, Barbara Ann Karmanos Cancer Institute, 4100 John R St., Detroit, MI, 48201, USA.
| |
Collapse
|
44
|
Shaul ME, Fridlender ZG. Cancer-related circulating and tumor-associated neutrophils - subtypes, sources and function. FEBS J 2018; 285:4316-4342. [PMID: 29851227 DOI: 10.1111/febs.14524] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 04/18/2018] [Accepted: 05/29/2018] [Indexed: 12/11/2022]
Abstract
In recent years, the role of neutrophils in cancer biology has been a matter of increasing interest. Many patients with advanced cancer show high levels of neutrophilia, tumor neutrophils are connected to dismal prognosis, and the neutrophil-to-lymphocyte ratio has been introduced as a significant prognostic factor for survival in many types of cancer. Neutrophils constitute an important portion of the infiltrating immune cells in the tumor microenvironment, but controversy has long surrounded the function of these cells in the context of cancer. Multiple evidences have shown that neutrophils recruited to the tumor can acquire either protumor or antitumor function. These findings have led to the identification of multiple and heterogeneous neutrophil subsets in the tumor and circulation. In addition, tumor-associated neutrophils (TANs) were shown to demonstrate functional plasticity, driven by multiple factors present in the tumor microenvironment. In this review, we examine the current knowledge on cancer-related circulating neutrophils, their source and the function of the different subtypes, both mature and immature. We then discuss the pro vs antitumor nature of TANs in cancer, their functional plasticity and the mechanisms that regulate neutrophil recruitment and polarization. Although the vast majority of the knowledge on neutrophils in cancer comes from murine studies, recent work has been done on human cancer-related neutrophils. In the final paragraphs, we expand on the current knowledge regarding the role of neutrophils in human cancer and examine the question whether cancer-related neutrophils (circulating or intratumoral) could be a new possible target for cancer immunotherapy.
Collapse
Affiliation(s)
- Merav E Shaul
- Institute of Pulmonary Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Zvi G Fridlender
- Institute of Pulmonary Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
45
|
Allosteric Effects between the Antibody Constant and Variable Regions: A Study of IgA Fc Mutations on Antigen Binding. Antibodies (Basel) 2018; 7:antib7020020. [PMID: 31544872 PMCID: PMC6698812 DOI: 10.3390/antib7020020] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/02/2018] [Accepted: 06/05/2018] [Indexed: 12/15/2022] Open
Abstract
Therapeutic antibodies have shifted the paradigm of disease treatments from small molecules to biologics, especially in cancer therapy. Despite the increasing number of antibody candidates, much remains unknown about the antibody and how its various regions interact. Recent findings showed that the antibody constant region can govern localization effects that are useful in reducing side effects due to systemic circulation by the commonly used IgG isotypes. Given their localized mucosal effects, IgA antibodies are increasingly promising therapeutic biologics. While the antibody Fc effector cell activity has been a focus point, recent research showed that the Fc could also influence antigen binding, challenging the conventional idea of region-specific antibody functions. To investigate this, we analysed the IgA antibody constant region and its distal effects on the antigen binding regions using recombinant Pertuzumab IgA1 and IgA2 variants. We found that mutations in the C-region reduced Her2 binding experimentally, and computational structural analysis showed that allosteric communications were highly dependent on the antibody hinge, providing strong evidence that we should consider antibodies as whole proteins rather than a sum of functional regions.
Collapse
|
46
|
Aliev TK, Dement’yeva IG, Toporova VA, Argentova VV, Pozdnyakova LP, Bokov MN, Votchitseva YA, Dolgikh DA, Varfolomeyev SD, Sveshnikov PG, Kirpichnikov MP. The Development and Study of Recombinant Immunoglobulin A to Hemagglutinins of the Influenza Virus. Acta Naturae 2018; 10:30-36. [PMID: 30116613 PMCID: PMC6087826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Indexed: 12/03/2022] Open
Abstract
We obtained recombinant variants of human antibody FI6 broadly specific to hemagglutinins of the influenza A virus. On the basis of a bi-promoter (CMV, hEF1-HTLV) vector, we developed genetic constructs for the expression of the heavy and light chains of the immunoglobulins of IgA1-, IgA2m1-, and IgG-isotypes. Following transfection and selection, stable Chinese hamster ovary (CHO) cell lines were produced. The antibodies of IgA1-, IgA2m1-, and IgG-isotypes were purified from culture media. We performed an immunochemical characterization and studied their interactions with influenza A strains of the H1N1- and H3N2-subtypes. It was shown that recombinant FI6 variants of the IgA-isotype retain the properties of the parental IgG antibody to demonstrate specificity to all the strains tested. The strongest binding was observed for the H1N1 subtype, which belongs to hemagglutinins of phylogenetic group I.
Collapse
Affiliation(s)
- T. K. Aliev
- Lomonosov Moscow State University, Department of Chemistry, Leninskie gory 1, bldg. 3, Moscow, 119991, Russia
| | - I. G. Dement’yeva
- Russian Research Center for Molecular Diagnostics and Therapy, Simferopolsky Blvd. 8, Moscow, 117149 , Russia
| | - V. A. Toporova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya Str. 16/10, Moscow, 117997, Russia
| | - V. V. Argentova
- Lomonosov Moscow State University, Faculty of Biology, Leninskie gory 1, bldg. 12, Moscow, 119991 , Russia
| | - L. P. Pozdnyakova
- Russian Research Center for Molecular Diagnostics and Therapy, Simferopolsky Blvd. 8, Moscow, 117149 , Russia
| | - M. N. Bokov
- Russian Research Center for Molecular Diagnostics and Therapy, Simferopolsky Blvd. 8, Moscow, 117149 , Russia
| | - Yu. A. Votchitseva
- Lomonosov Moscow State University, Faculty of Biology, Leninskie gory 1, bldg. 12, Moscow, 119991 , Russia
| | - D. A. Dolgikh
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya Str. 16/10, Moscow, 117997, Russia
- Lomonosov Moscow State University, Faculty of Biology, Leninskie gory 1, bldg. 12, Moscow, 119991 , Russia
| | - S. D. Varfolomeyev
- Lomonosov Moscow State University, Department of Chemistry, Leninskie gory 1, bldg. 3, Moscow, 119991, Russia
| | - P. G. Sveshnikov
- Russian Research Center for Molecular Diagnostics and Therapy, Simferopolsky Blvd. 8, Moscow, 117149 , Russia
| | - M. P. Kirpichnikov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya Str. 16/10, Moscow, 117997, Russia
- Lomonosov Moscow State University, Faculty of Biology, Leninskie gory 1, bldg. 12, Moscow, 119991 , Russia
| |
Collapse
|
47
|
Aliev TK, Dement'eva IG, Toporova VA, Argentova VV, Pozdnyakova LP, Bokov MN, Dolgikh DA, Sveshnikov PG, Kirpichnikov MP. The Development and Inmmunochemical Properties of the Dimer of Immunoglobulin A Specific to the Influenza Virus A Hemagglutinin. DOKL BIOCHEM BIOPHYS 2018. [PMID: 29536306 DOI: 10.1134/s160767291801009x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
We obtained dimeric forms of IgA1- and IgA2m1-isotypes of FI6 antibody broadly specific to hemagglutinins of different subtypes of influenza A virus. It was shown that the dimers of IgA1 isotype are characterized by a higher antigen-binding activity compared to the IgA2m1 dimers. The affinity of IgA1 dimers to the strains of the H1N1 subtype is higher than that of the H3N2 subtype, which correlates with the properties of the parental human FI6 antibody.
Collapse
Affiliation(s)
- T K Aliev
- Faculty of Biology, Moscow State University, Moscow, 119991, Russia.
| | - I G Dement'eva
- All-Russia Research Center of Molecular Diagnostics and Therapy, Moscow, 117149, Russia
| | - V A Toporova
- All-Russia Research Center of Molecular Diagnostics and Therapy, Moscow, 117149, Russia
| | - V V Argentova
- Faculty of Biology, Moscow State University, Moscow, 119991, Russia
| | - L P Pozdnyakova
- All-Russia Research Center of Molecular Diagnostics and Therapy, Moscow, 117149, Russia
| | - M N Bokov
- All-Russia Research Center of Molecular Diagnostics and Therapy, Moscow, 117149, Russia
| | - D A Dolgikh
- Faculty of Biology, Moscow State University, Moscow, 119991, Russia.,Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - P G Sveshnikov
- All-Russia Research Center of Molecular Diagnostics and Therapy, Moscow, 117149, Russia
| | - M P Kirpichnikov
- Faculty of Biology, Moscow State University, Moscow, 119991, Russia.,Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| |
Collapse
|
48
|
Capodicasa C, Catellani M, Moscetti I, Bromuro C, Chiani P, Torosantucci A, Benvenuto E. Comparative analysis of plant-produced, recombinant dimeric IgA against cell wall β-glucan of pathogenic fungi. Biotechnol Bioeng 2017; 114:2729-2738. [PMID: 28832951 DOI: 10.1002/bit.26403] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/30/2017] [Accepted: 08/17/2017] [Indexed: 12/14/2022]
Abstract
Immunoglobulins A (IgA) are crucially involved in protection of human mucosal surfaces from microbial pathogens. In this work, we devised and expressed in plants recombinant chimeric antifungal antibodies (Abs) of isotype A (IgA1, IgA2, and scFvFcA1), derived from a murine mAb directed to the fungal cell wall polysaccharide β-glucan which had proven able to confer protection against multiple pathogenic fungi. All recombinant IgA (rIgA) were expressed and correctly assembled in dimeric form in plants and evaluated for yield, antigen-binding efficiency and antifungal properties in vitro, in comparison with a chimeric IgG1 version. Production yields and binding efficiency to purified β-glucans showed significant variations not only between Abs of different isotypes but also between the different IgA formats. Moreover, only the dimeric IgA1 was able to strongly bind cells of the fungal pathogen Candida albicans and to restrain its adhesion to human epithelial cells. Our data indicate that IgG to IgA switch and differences in molecular structure among different rIgA formats can impact expression in plant and biological activity of anti-β-glucans Abs and provide new insights for the design of recombinant IgA as anti-infective immunotherapeutics, whose potential is still poorly investigated.
Collapse
Affiliation(s)
- Cristina Capodicasa
- Laboratory of Biotechnology, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Research Center Casaccia, Rome, Italy
| | - Marcello Catellani
- Laboratory of Biotechnology, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Research Center Casaccia, Rome, Italy
| | - Ilaria Moscetti
- Laboratory of Biotechnology, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Research Center Casaccia, Rome, Italy
| | - Carla Bromuro
- Department of Infectious, Parasitic and Immune-Mediated Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Paola Chiani
- Department of Infectious, Parasitic and Immune-Mediated Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Antonella Torosantucci
- Department of Infectious, Parasitic and Immune-Mediated Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Eugenio Benvenuto
- Laboratory of Biotechnology, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Research Center Casaccia, Rome, Italy
| |
Collapse
|
49
|
Treffers LW, Hiemstra IH, Kuijpers TW, van den Berg TK, Matlung HL. Neutrophils in cancer. Immunol Rev 2017; 273:312-28. [PMID: 27558343 DOI: 10.1111/imr.12444] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Neutrophils play an important role in cancer. This does not only relate to the well-established prognostic value of the presence of neutrophils, either in the blood or in tumor tissue, in the context of cancer progression or for the monitoring of therapy, but also to their active role in the progression of cancer. In the current review, we describe what is known in general about the role of neutrophils in cancer. What is emerging is a complex, rather heterogeneous picture with both pro- and anti-tumorigenic roles, which apparently differs with cancer type and disease stage. Furthermore, we will discuss the well-known role of neutrophils as myeloid-derived suppressor cells (MDSC), and also on the role of neutrophils as important effector cells during antibody therapy in cancer. It is clear that neutrophils contribute substantially to cancer progression in multiple ways, and this includes both direct effects on the cancer cells and indirect effect on the tumor microenvironment. While in many cases neutrophils have been shown to promote tumor progression, for instance by acting as MDSC, there are also protective effects, particularly when antibody immunotherapy is performed. A better understanding of the role of neutrophils is likely to provide opportunities for immunomodulation and for improving the treatment of cancer patients.
Collapse
Affiliation(s)
- Louise W Treffers
- Sanquin Research, and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Ida H Hiemstra
- Sanquin Research, and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Taco W Kuijpers
- Sanquin Research, and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Timo K van den Berg
- Sanquin Research, and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Hanke L Matlung
- Sanquin Research, and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
50
|
Epp A, Sullivan KC, Herr AB, Strait RT. Immunoglobulin Glycosylation Effects in Allergy and Immunity. Curr Allergy Asthma Rep 2017; 16:79. [PMID: 27796794 DOI: 10.1007/s11882-016-0658-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW The aim of this review will be to familiarize the reader with the general area of antibody (Ab) glycosylation and to summarize the known functional roles of glycosylation and how glycan structure can contribute to various disease states with emphasis on allergic disease. RECENT FINDINGS Both immunoglobulin (Ig) isotype and conserved Fc glycosylation sites often dictate the downstream activity of an Ab where complexity and degree of glycosylation contribute to its ability to bind Fc receptors (FcRs) and activate complement. Most information on the effects of glycosylation center on IgG in cancer therapy and autoimmunity. In cancer therapy, glycosylation modifications that enhance affinity for activating FcRs are utilized to facilitate immune-mediated tumor cell killing. In autoimmunity, disease severity has been linked to alterations in the presence, location, and composition of Fc glycans. Significantly less is understood about the role of glycosylation in the setting of allergy and asthma. However, recent data demonstrate that glycosylation of IgE at the asparagine-394 site of Cε3 is necessary for IgE interaction with the high affinity IgE receptor but, surprisingly, glycosylation has no effect on IgE interaction with its low-affinity lectin receptor, CD23. Variations in the specific glycoform may modulate the interaction of an Ig with its receptors. Significantly more is known about the functional effects of glycosylation of IgG than for other Ig isotypes. Thus, the role of glycosylation is much better understood in the areas of autoimmunity and cancer therapy, where IgG is the dominant isotype, than in the field of allergy, where IgE predominates. Further work is needed to fully understand the role of glycan variation in IgE and other Ig isotypes with regard to the inhibition or mediation of allergic disease.
Collapse
Affiliation(s)
- Alexandra Epp
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Kathryn C Sullivan
- Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Andrew B Herr
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Richard T Strait
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA. .,Division of Emergency Medicine, Cincinnati Children's Hospital, 3333 Burnet Ave, ML 2008, Cincinnati, OH, 45229, USA.
| |
Collapse
|